US20080268052A1 - Collagen preparation and method of isolation - Google Patents

Collagen preparation and method of isolation Download PDF

Info

Publication number
US20080268052A1
US20080268052A1 US11/903,326 US90332607A US2008268052A1 US 20080268052 A1 US20080268052 A1 US 20080268052A1 US 90332607 A US90332607 A US 90332607A US 2008268052 A1 US2008268052 A1 US 2008268052A1
Authority
US
United States
Prior art keywords
collagen
oligomers
isolated
engineered
matrix
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US11/903,326
Other versions
US8084055B2 (en
Inventor
Sherry L. Voytik-Harbin
Seth Kreger
Brett Bell
Jennifer Bailey
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Purdue Research Foundation
Original Assignee
Voytik-Harbin Sherry L
Seth Kreger
Brett Bell
Jennifer Bailey
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Voytik-Harbin Sherry L, Seth Kreger, Brett Bell, Jennifer Bailey filed Critical Voytik-Harbin Sherry L
Priority to US11/903,326 priority Critical patent/US8084055B2/en
Publication of US20080268052A1 publication Critical patent/US20080268052A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT EXECUTIVE ORDER 9424, CONFIRMATORY LICENSE Assignors: PURDUE UNIVERSITY
Priority to US13/331,483 priority patent/US8512756B2/en
Application granted granted Critical
Publication of US8084055B2 publication Critical patent/US8084055B2/en
Assigned to PURDUE RESEARCH FOUNDATION reassignment PURDUE RESEARCH FOUNDATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VOYTIK-HARBIN, SHERRY L., BELL, BRETT, BAILEY, JENNIFER, KREGER, SETH
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/14Macromolecular materials
    • A61L27/22Polypeptides or derivatives thereof, e.g. degradation products
    • A61L27/24Collagen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/78Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin, cold insoluble globulin [CIG]
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08LCOMPOSITIONS OF MACROMOLECULAR COMPOUNDS
    • C08L89/00Compositions of proteins; Compositions of derivatives thereof
    • C08L89/04Products derived from waste materials, e.g. horn, hoof or hair
    • C08L89/06Products derived from waste materials, e.g. horn, hoof or hair derived from leather or skin, e.g. gelatin

Definitions

  • This invention relates to collagen compositions, methods for preparing those collagen compositions, and graft compositions formed from those collagen compositions. More particularly, the invention relates to methods of isolating collagen that exhibits an enhanced rate of polymerization and enhanced micro-structural and mechanical properties upon polymerization and to such collagen compositions and graft compositions formed from such collagen compositions.
  • ECM extracellular matrix
  • the extracellular matrix (ECM) as it occurs in vivo plays a crucial role in the organization, homeostasis, and function of tissues and organs.
  • Continuous communication between cells and their surrounding ECM environment orchestrates critical processes such as the acquisition and maintenance of differentiated phenotypes during embryogenesis, the development of form (morphogenesis), angiogenesis, wound healing, and even tumor metastasis.
  • morphogenesis the development of form
  • angiogenesis angiogenesis
  • Both biochemical and biophysical signals from the ECM modulate fundamental cellular activities including adhesion, migration, proliferation, differential gene expression, and programmed cell death.
  • the realization of the significance of the ECM to the organization, homeostasis, and function of tissues and organs has led to a renewed interest in characterizing ECM constituents and the relationship of these constituents to the functioning of the ECM.
  • ECMs including various basement membrane tissues and other extracellular matrix tissues obtained from natural sources and matrices formed from isolated ECM components, can be utilized as tissue graft compositions for remodeling tissues in vivo or for in vitro applications.
  • Complex scaffolds representing combinations of isolated extracellular matrix components in a natural or processed form are commercially available and can be used as tissue graft compositions (e.g., Human Extracellular Matrix (Becton Dickinson) and MATRIGEL®).
  • tissue graft compositions e.g., Human Extracellular Matrix (Becton Dickinson) and MATRIGEL®.
  • Basement membrane tissues and other extracellular matrix tissues such as tissue material derived from submucosal tissues, harvested from warm-blooded vertebrates have also shown great promise as unique graft materials for inducing the repair of damaged or diseased tissues in vivo, and for inducing the proliferation of cells in vitro.
  • purified collagen can be used to produce an engineered ECM material prepared under conditions that regulate the polymerization of collagen in a controlled manner. This result is more difficult to achieve with existing intact or processed ECMs from natural sources and with ECMs and collagen preparations from commercial sources.
  • modifying the conditions used to isolate collagen results in collagen preparations with properties that enhance the rate at which the collagen polymerizes and that enhance the microstructural and mechanical properties of the collagen upon polymerization (e.g., the mechanical integrity of the engineered ECM that is formed upon collagen polymerization).
  • the methods of collagen isolation and the collagen compositions described herein allow for the controlled alteration of the microstructural and subsequent mechanical properties of a resulting engineered ECM for such uses as graft compositions for inducing the repair of damaged or diseased tissues in vivo, and for inducing the proliferation and other fundamental behaviors of cells in vitro.
  • a method for isolating collagen comprises the steps of obtaining a collagen-containing source material, comminuting the source material, mixing the comminuted source material with an extraction solution, extracting the comminuted source material to form a soluble fraction and an insoluble fraction, obtaining the insoluble fraction, extracting the collagen from the insoluble fraction to form a soluble collagen fraction, precipitating the collagen from the soluble collagen fraction, and resuspending the precipitate in an aqueous solution wherein the aqueous solution used to resuspend the collagen precipitate is an acidic solution.
  • An isolated collagen composition prepared by this method is also provided.
  • the collagen-containing source material is selected from the group consisting of placental tissue, bladder tissue, intestinal tissue, alimentary tract tissue, ovarian tissue, pericardial tissue, animal tail tissue, liver tissue, skin tissue, and any other suitable collagen-containing source material
  • the collagen-containing source material is selected from the group consisting of bladder tissue and skin tissue
  • the collagen-containing source material is porcine skin tissue
  • the source material is frozen in liquid nitrogen prior to the comminuting step
  • the source material is frozen in liquid nitrogen during the comminuting step
  • the source material is frozen in liquid nitrogen prior to and during the comminuting step
  • the source material is frozen at a temperature of ⁇ 20° C.
  • the source material is frozen at a temperature of ⁇ 40° C. or below prior to or during the comminuting step, 9) the source material is frozen at a temperature of ⁇ 60° C. or below prior to or during the comminuting step, 10) the source material is frozen at a temperature of ⁇ 80° C.
  • the mixing step is performed by blending or stirring, 12) the mixing step is performed by stirring, 13) the soluble collagen fraction is not filtered between the step of extracting the collagen from the insoluble fraction to form the soluble collagen fraction and the step of precipitating the collagen from the soluble collagen fraction, 14) the method further comprises the step of lyophilizing the collagen precipitate, 15) the method further comprises the step of polymerizing the collagen prior to or after lyophilization, 16) the collagen is precipitated by dialysis against a buffered solution, and 17) the dialysis tubing has a molecular weight cut-off of about 12,000 to about 14,000. In an alternative embodiment, any of these steps can be used in any combination.
  • a method for engineering matrices with enhanced polymerization characteristics comprises the steps of obtaining collagen oligomers, polymerizing the collagen oligomers, and forming the engineered matrices with enhanced polymerization characteristics.
  • An engineered matrix prepared by this method is also provided.
  • the enhanced polymerization characteristics are selected from the group consisting of an enhanced rate of polymerization, a reduced lag time for polymerization, and an enhanced mechanical integrity
  • the enhanced mechanical integrity is selected from the group consisting of strength and stiffness.
  • the collagen oligomers polymerize with a maximal t 1 ⁇ 2 selected from the group consisting of about 3 minutes, about 2.5 minutes, about 2.0 minutes, about 1.5 minutes, and about 1 minute.
  • the collagen oligomers are obtained by isolation from a collagen-containing source material that is a tissue naturally enriched with collagen oligomers, the collagen oligomers are obtained by isolating and then chemically cross-linking collagen, or the collagen oligomers are isolated from a diseased tissue or a genetically-modified tissue.
  • an engineered matrix comprises a predetermined percentage of collagen oligomers based on total isolated collagen used to make the engineered matrix.
  • the collagen is isolated from a tissue naturally enriched with collagen oligomers, the collagen oligomers are isolated from a diseased tissue or a genetically-modified tissue, or the collagen oligomers are obtained by isolating and then chemically cross-linking collagen.
  • the predetermined percentage of collagen oligomers is selected from the group consisting of about 0.5% to about 100%, about 1.0% to about 100%, about 2% to about 100%, about 3% to about 100%, about 5% to about 100%, about 10% to about 100%, about 15% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 90% to about 100%, about 95% to about 100%, and about 100%.
  • the engineered matrix further comprises a predetermined percentage of isolated collagen monomers.
  • a graft composition comprises an engineered matrix comprising collagen oligomers wherein the matrix has a predetermined percentage of collagen oligomers based on total isolated collagen used to make the engineered matrix.
  • the predetermined percentage of collagen oligomers is selected from the group consisting of about 0.5% to about 100%, about 1.0% to about 100%, about 2% to about 100%, about 3% to about 100%, about 5% to about 100%, about 10% to about 100%, about 15% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 90% to about 100%, about 95% to about 100%, and about 100%.
  • the collagen oligomers are isolated from a diseased tissue or a genetically-modified tissue, the collagen oligomers are obtained by isolating and then chemically cross-linking collagen, or the collagen oligomers are isolated from a natural tissue enriched with collagen oligomers.
  • the graft composition further comprises a predetermined percentage of collagen monomers based on total isolated collagen used to make the engineered matrix.
  • the inclusion of increased amounts of collagen oligomers in a collagen composition may increase the rate of polymerization, facilitate hierarchical assembly of component collagen fibrils, and enhance mechanical properties (e.g., strength and stiffness).
  • FIG. 1 shows a polyacrylamide gel electrophoresis comparison of collagen isolated from pig skin, using the methods described herein, and a commercially available collagen preparation.
  • FIG. 2 shows a comparison by using turbidity analysis of the kinetics of polymerization of collagen isolated from pig skin using the methods described herein and a commercially available collagen preparation.
  • FIG. 3 shows a comparison using confocal reflection microscopy of the microstructure of an engineered ECM prepared from pig skin collagen, using the methods described herein, and a commercially available collagen preparation.
  • FIG. 4 shows a comparison using scanning electron microscopy of the microstructure of an engineered ECM prepared from pig skin collagen, using the methods described herein, and a commercially available collagen preparation.
  • FIG. 5 shows a comparison using a compression test of the mechanical properties (e.g., strength and modulus (stiffness)) of an engineered ECM prepared from pig skin collagen, using the methods described herein, and a commercially available collagen preparation.
  • mechanical properties e.g., strength and modulus (stiffness)
  • FIG. 6 shows a polyacrylamide gel electrophoresis comparison of cyanogen bromide peptides from collagen isolated from pig skin collagen, using the methods described herein, and commercially available collagen preparations.
  • FIG. 7 shows the shear storage (G′) modulus during polymerization of an engineered ECM prepared from pig skin collagen, using the methods described herein, compared to a commercially available collagen preparation.
  • FIGS. 8A and B show the growth of mesenchymal stem cells on an engineered collagen ECM polymerized at 0.5 mg/ml ( FIG. 8A ) or 3 mg/ml ( FIG. 8B ) of collagen, and polymerized from pig skin collagen, isolated according to the methods described herein.
  • FIG. 9 shows the time-dependent glycerol-based separation of pig skin collagen type I oligomers and monomers. At each time point, the supernatant and pellet represent the monomer-rich and oligomer-rich fractions, respectively.
  • FIGS. 10A and B show glycerol-based separation of pig skin collagen type I oligomers and monomers.
  • Pig skin collagen was polymerized in the presence of glycerol for 7 days. The mixture was centrifuged and the supernatant and pellet retained, dialyzed against dilute acetic acid and lyophilized to dryness. The supernatant and pellet represented monomer- and oligomer-rich collagen compositions, respectively, as determined by SDS-PAGE (4%).
  • FIG. 10A shows the SDS-PAGE gel and FIG. 10B shows the corresponding densitometry analysis.
  • FIG. 11 shows SDS-PAGE (12%) analysis of CNBr peptide maps of collagen preps in which oligomer:monomer content is varied. Upon CNBr digestion, collagen preps yield distinct peptide fingerprints indicating distinct molecular compositions and cross-linked moieties.
  • FIGS. 12 A-B show the oligomer:monomer content of pig skin collagen preparation affects polymerization (self-assembly) kinetics.
  • An increase in oligomer content decreases lag time ( FIG. 12 B) and increases the rate of polymerization ( FIG. 12 A).
  • FIG. 12A shows A 405 vs. time.
  • FIG. 13 shows the comparison of mechanical behavior of 3D engineered ECMs prepared with pig skin collagen preps of varied monomer:oligomer content.
  • the shear storage modulus (G′) provides a measure of the stiffness of solid (elastic) components. Results show that G′ or stiffness of matrices increases with oligomer content.
  • FIG. 14 shows the comparison of mechanical behavior of 3D engineered ECMs prepared with pig skin collagen preps of varied monomer:oligomer content. An increase in delta as a function of strain as observed with the monomer-rich preparation indicates more fluid-like (viscous) behavior.
  • FIG. 15 shows the comparison of mechanical behavior of 3D engineered ECMs prepared with pig skin collagen preps of varied monomer:oligomer content. Matrices produced with collagen preps consisting of higher oligomer content show increased compressive modulus (stiffness) and failure strength.
  • FIG. 16 shows the compressive modulus (stiffness) values as a function of the oligomer:monomer content of the pig skin collagen preparation.
  • FIG. 17 shows the effects of oligomer content on the 3D collagen fibril microstructure of engineered ECMs.
  • FIG. 18 shows the effects of oligomer content on the 3D collagen fibril microstructure of engineered ECMs.
  • FIG. 19 shows the compositional analysis of different collagen preparations as determined using SDS-PAGE (4%). Results show that the pig skin collagen preparation uniquely contains collagen oligomers that run intermediate between commonly identified dimeric ( ⁇ ) and trimeric ( ⁇ ) forms of collagen. Upon pepsin treatment of pig skin collagen, the content of unique collagen oligomers as other higher molecular weight collagen species is significantly reduced.
  • FIG. 20 shows the kinetics of polymerization for the different collagen preparations as determined using spectrophotometric-based turbidity assay. Results show that the polymerization kinetics of collagen is highly dependent upon the collagen preparation. More specifically, polymerization of pig skin collagen shows a decreased lag phase and an increased rate of polymerization (slope of growth phase).
  • FIG. 21 shows the comparison of final absorbance (405 nm) values for different collagen preparations as determined from spectrophometric based turbidity assay. Results represent average of last 10 absorbance values.
  • FIG. 22 shows the collagen fibril microstructure of 3D engineered ECMs prepared with different purified type I collagen sources. Each collagen preparation was polymerized at two different concentrations (0.5 mg/ml and 2 mg/ml) using the same reaction conditions. Images represent 2D projections of 3D image stacks as collected using confocal reflection microscopy. Results show that the collagen fibril microstructure of 3D engineered ECMs is highly dependent upon the collagen preparation.
  • FIG. 23 shows the comparison of mechanical behavior 3D engineered ECMs prepared with different collagen preparations and tested in oscillatory shear.
  • the storage modulus (G′) provides a measure of the stiffness of the solid (elastic) components. Results show that G′ or stiffness of matrices prepared with pig skin collagen are significantly greater than those obtained with the commercial collagen preparations (PureCol and Sigma).
  • FIGS. 24A-B show a comparison of collagen sources.
  • FIG. 24A shows microstructural analysis of engineered ECMs using confocal reflection microscopy (fibril density vs. collagen concentration).
  • FIG. 24B shows microstructural analysis of engineered ECMs using confocal reflection microscopy (average fibril diameter vs. collagen concentration).
  • lyophilized means that water is removed from the composition, typically by freeze-drying under a vacuum. However, lyophilization can be performed by any method known to the skilled artisan and the method is not limited to freeze-drying under a vacuum.
  • collagen-based matrix means a matrix that comprises collagen.
  • the “collagen-based matrices” described herein can be engineered from isolated collagen.
  • engineered matrix means a collagen-based matrix that is polymerized under conditions that are systematically varied where the conditions are selected from the group consisting of, but not limited to, pH, phosphate concentration, temperature, buffer composition, ionic strength, and composition and concentration of collagen.
  • isolated collagen means any type of collagen, naturally present in a collagen-containing source material (described below) wherein the collagen has been at least partially purified by isolation and removal from the collagen-containing source material.
  • sterilization or “sterilize” or “sterilized” means removing unwanted contaminants including, but not limited to, endotoxins, nucleic acid contaminants, and infectious agents.
  • the present invention relates to a method of preparing a collagen-based matrix.
  • methods for isolating collagen and then preparing the collagen-based matrix are provided.
  • the method for isolating collagen comprises the steps of obtaining a collagen-containing source material, comminuting the source material, mixing the comminuted source material with an extraction solution, extracting the comminuted source material to form a soluble fraction and an insoluble fraction, obtaining the insoluble fraction, extracting the collagen from the insoluble fraction to form a soluble collagen fraction, precipitating the collagen from the soluble collagen fraction, and resuspending the precipitate in an aqueous solution wherein the aqueous solution used to resuspend the collagen precipitate is an acidic solution.
  • the isolated collagen can be precipitated by dialysis against a buffered solution, for example, using dialysis tubing with a molecular weight cut-off of about 12,000 to about 14,000.
  • the isolated collagen can be lyophilized after precipitation.
  • a polymerizing step can be performed under conditions that are systematically varied where the conditions are selected from the group consisting of pH, phosphate concentration, temperature, buffer composition, ionic strength, the specific isolated collagen components present, and the concentration of the isolated collagen components present.
  • the isolated collagen can be lyophilized prior to polymerization.
  • the isolated collagen can be lyophilized in an acid, such as acetic acid, hydrochloric acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid.
  • the isolated collagen can be polymerized after precipitation, and optionally, the engineered matrix that results can be lyophilized.
  • the invention relates to a collagen-based matrix prepared using the methods described above.
  • the collagen-based matrix can contain fibrils with specific characteristics, including, but not limited to, a fibril area fraction or a fibril volume fraction (i.e., density) of about 0.1% to about 100%, about 0.5% to about 100%, about 0.5% to about 26%, about 1% to about 100%, about 1% to about 26%, about 1% to about 7%, about 1% to about 15%, about 7% to about 26%, about 20% to about 30%, about 20% to about 50%, about 20% to about 70%, and about 20% to about 100%, about 30% to about 50%, about 30% to about 70%, and about 30% to about 100% and/or a modulus (e.g., an elastic or linear modulus, a compressive modulus, or a shear storage modulus) of about 0.5 kPa to about 40 kPa, about 30 kPa to 100 kPa, about 30 kPa to about 1000 kPa
  • a modulus e.g.
  • Exemplary of tissues useful as a collagen-containing source material for isolating collagen to make the collagen-based matrices described herein are submucosa tissues or any other extracellular matrix-containing tissues of a warm-blooded vertebrate. Exemplary methods of preparing submucosa tissues are described in U.S. Pat. Nos. 4,902,508; 5,281,422; and 5,275,826, each incorporated herein by reference. Extracellular matrix material-containing tissues other than submucosa tissue may be used in accordance with the methods and compositions described herein. Methods of preparing other extracellular matrix material-derived tissues are known to those skilled in the art. For example, see U.S. Pat. Nos.
  • the collagen-containing source material can be selected from the group consisting of placental tissue, ovarian tissue, animal tail tissue, and skin tissue (e.g., see Example 1 and Gallop, et al., Preparation and Properties of Soluble Collagens, Meth. Enzymol. 6: 635-641 (1963), incorporated herein by reference). Any suitable extracellular matrix-containing tissue can be used as a collagen-containing source material.
  • the extracellular matrix material-derived tissues used as the collagen-containing source material can be derived from a vertebrate tissue material comprising submucosa.
  • vertebrate tissue material comprising submucosa can be obtained from intestinal tissue harvested from animals raised for meat production, including, for example, pigs, cattle and sheep or other warm-blooded vertebrates.
  • the tissue material comprising submucosa can be obtained from intestine, stomach, urinary bladder, the uterus, and any other submucosa-containing tissue.
  • tissue material comprising submucosa is described in U.S. Pat. No. 4,902,508, the disclosure of which is incorporated herein by reference.
  • a segment of vertebrate intestine for example, preferably harvested from porcine, ovine or bovine species, but not excluding other species, is subjected to abrasion using a longitudinal wiping motion to remove cells.
  • the tissue material comprising submucosa is rinsed under hypotonic conditions, such as with water or with saline under hypotonic conditions and is optionally sterilized.
  • compositions can be prepared by mechanically removing the luminal portion of the tunica mucosa and the external muscle layers and/or lysing resident cells with hypotonic washes, such as water or saline under hypotonic conditions.
  • hypotonic washes such as water or saline under hypotonic conditions.
  • the tissue material comprising submucosa can be stored in a hydrated or dehydrated state prior to extraction.
  • the tissue material comprising submucosa can comprise any delamination embodiment, including the tunica submucosa delaminated from both the tunica muscularis and at least the luminal portion of the tunica mucosa of a warm-blooded vertebrate.
  • the isolated collagen can also contain glycoproteins, proteoglycans, glycosaminoglycans (e.g., chondroitins and heparins), etc. extracted from the insoluble fraction with the collagen.
  • the engineered matrices prepared by the methods described herein can serve as matrices for the regrowth of endogenous tissues at the implantation site (e.g., biological remodeling) which can assume the characterizing features of the tissue(s) with which they are associated at the site of implantation, insertion, or injection.
  • the collagen-containing source material, the isolated collagen, or the collagen-based matrix, including an engineered matrix can be disinfected and/or sterilized using conventional sterilization techniques including glutaraldehyde tanning, formaldehyde tanning at acidic pH, propylene oxide or ethylene oxide treatment, gas plasma sterilization, gamma radiation, electron beam, and/or peracetic acid sterilization. Sterilization techniques which do not adversely affect the structure and biotropic properties of the source material or the collagen can be used.
  • Illustrative sterilization techniques are exposing the collagen-containing source material, the isolated collagen, or the collagen-based matrix, including an engineered matrix, to peracetic acid, 1-4 Mrads gamma irradiation (or 1-2.5 Mrads of gamma irradiation), ethylene oxide treatment, or gas plasma sterilization.
  • peracetic acid can be used for sterilization.
  • the collagen-containing source material is comminuted by tearing, cutting, grinding, or shearing the collagen-containing source material.
  • the collagen-containing source material can be comminuted by shearing in a high-speed blender, or by grinding the collagen-containing source material in a frozen state (e.g., at a temperature of ⁇ 20° C., ⁇ 40° C., ⁇ 60° C., or ⁇ 80° C. or below prior to or during the comminuting step) and then lyophilizing the material to produce a powder having particles ranging in size from about 0.1 mm 2 to about 1.0 mm 2 .
  • the collagen-containing source material is comminuted by freezing and pulverizing under liquid nitrogen in an industrial blender.
  • the collagen-containing source material can be frozen in liquid nitrogen prior to, during, or prior to and during the comminuting step.
  • the material is mixed (e.g., by blending or stirring) with an extraction solution to extract and remove soluble proteins.
  • extraction solutions include sodium acetate (e.g., 0.5 M and 1.0 M).
  • Other exemplary methods for extracting soluble proteins are known to those skilled in the art and are described in detail in U.S. Pat. No. 6,375,989, incorporated herein by reference.
  • Illustrative extraction excipients include, for example, chaotropic agents such as urea, guanidine, sodium chloride or other neutral salt solutions, magnesium chloride, and non-ionic or ionic surfactants.
  • the soluble fraction can be separated from the insoluble fraction to obtain the insoluble fraction.
  • the insoluble fraction can be separated from the soluble fraction by centrifugation (e.g., 2000 rpm at 4° C. for 1 hour). In alternative embodiments, other separation techniques known to those skilled in the art, such as filtration, can be used.
  • the initial extraction step can be repeated one or more times, discarding the soluble fractions.
  • one or more steps can be performed of washing with water the insoluble fraction, followed by centrifugation, and discarding of the supernatant where the water is the supernatant.
  • the insoluble fraction can then be extracted (e.g., with 0.075 M sodium citrate) to obtain the isolated collagen.
  • the extraction step can be repeated multiple times retaining the soluble fractions.
  • the accumulated soluble fractions can be combined and can be clarified to form the soluble fraction, for example by centrifugation (e.g., 2000 rpm at 4° C. for 1 hour).
  • the soluble fraction can be fractionated to precipitate the isolated collagen.
  • the soluble fraction can be fractionated by dialysis.
  • Exemplary molecular weight cut-offs for the dialysis tubing or membrane are from about 3,500 to about 12,000 or about 3,500 to about 5,000 or about 12,000 to about 14,000.
  • the fractionation for example by dialysis, can be performed at about 2° C. to about 37° C. for about 1 hour to about 96 hours.
  • the soluble fraction is dialyzed against a buffered solution (e.g., 0.02 M sodium phosphate dibasic).
  • the fractionation can be performed at any temperature, for any length of time, and against any suitable buffered solution.
  • the precipitated collagen is then collected by centrifugation (e.g., 2000 rpm at 4° C. for 1 hour).
  • one or more steps can be performed of washing the precipitate with water, followed by centrifugation, and discarding of the supernatant where the water is the supernatant.
  • the precipitated collagen can then be resuspended in an aqueous solution wherein the aqueous solution is acidic.
  • the aqueous acidic solution can be an acetic acid solution, but any other acids including hydrochloric acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid can be used.
  • acids at concentrations of from about 0.001 N to about 0.1 N, from about 0.005 N to about 0.1 N, from about 0.01 N to about 0.1 N, from about 0.05 N to about 0.1 N, from about 0.001 N to about 0.05 N, from about 0.001 N to about 0.01 N, or from about 0.01 N to about 0.05 N can be used to resuspend the precipitate.
  • the term “lyophilized” means that water is removed from the composition, typically by freeze-drying under a vacuum.
  • the isolated resuspended collagen can be lyophilized after it is resuspended.
  • the polymerized matrix itself can be lyophilized.
  • the resuspended collagen is first frozen, and then placed under a vacuum.
  • the resuspended collagen can be freeze-dried under a vacuum.
  • the precipitated collagen can be lyophilized before resuspension. Any method of lyophilization known to the skilled artisan can be used.
  • the acids described above can be used as adjuvants for storage after lyophilization in any combination.
  • the acids that can be used as adjuvants for storage include hydrochloric acid, acetic acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid, and these acids can be used at any of the above-described concentrations.
  • the lyophilizate can be stored (e.g., lyophilized in and stored in) an acid, such as acetic acid, at a concentration of from about 0.001 N to about 0.5 N or from about 0.01 N to about 0.5 N.
  • the lyophilizate can be stored in water with a pH of about 6 or below.
  • the lyophilized product can be stored dry.
  • lyoprotectants, cryoprotectants, lyophilization accelerators, or crystallizing excipients e.g., ethanol, isopropanol, mannitol, trehalose, maltose, sucrose, tert-butanol, and tween 20
  • crystallizing excipients e.g., ethanol, isopropanol, mannitol, trehalose, maltose, sucrose, tert-butanol, and tween 20
  • the resuspended collagen is sterilized.
  • Exemplary sterilizing and/or disinfecting agents are described above, but any sterilizing and/or disinfecting agent or method of sterilization known in the art can be used.
  • the resuspended collagen can be sterilized using chloroform, glutaraldehyde, formaldehyde, acidic pH, propylene oxide, ethylene oxide, gas plasma sterilization, gamma radiation, electron beam sterilization, or peracetic acid sterilization, or combinations thereof, and the like.
  • Illustrative sterilization techniques are exposing the resuspended collagen to peracetic acid, 1-4 Mrads gamma irradiation (or 1-2.5 Mrads of gamma irradiation), ethylene oxide treatment, or gas plasma sterilization.
  • the isolated collagen can be sterilized before lyophilization.
  • the isolated collagen can be sterilized after lyophilization or the collagen-containing source material can be sterilized. Sterilization of the collagen-containing source material can be performed, for example, as described in U.S. Pat. Nos. 4,902,508 and 6,206,931, incorporated herein by reference.
  • the polymerized matrix formed from the isolated collagen is sterilized.
  • the isolated collagen is directly sterilized after resuspension, for example, with peracetic acid or with peracetic acid and ethanol (e.g., by the addition of 0.18% peracetic acid and 4.8% ethanol to the resuspended collagen solution before lyophilization).
  • sterilization can be carried out during the fractionation step.
  • the isolated collagen composition can be dialyzed against chloroform, peracetic acid, or a solution of peracetic acid and ethanol to disinfect or sterilize the isolated collagen.
  • the isolated collagen can be sterilized by dialysis against a solution of peracetic acid and ethanol (e.g., 0.18% peracetic acid and 4.8% ethanol).
  • the chloroform, peracetic acid, or peracetic acid/ethanol can be removed prior to lyophilization, for example by dialysis against an acid, such as 0.01 N acetic acid.
  • the lyophilized composition can be sterilized directly after rehydration, for example, by the addition of 0.18% peracetic acid and 4.8% ethanol.
  • the sterilizing agent can be removed prior to polymerization of the isolated collagen to form fibrils.
  • the lyophilized composition can be stored frozen, refrigerated, or at room temperature (for example, at about ⁇ 80° C. to about 25° C.). Storage temperatures are selected to stabilize the collagen.
  • the compositions can be stored for about 1-26 weeks, or longer.
  • the isolated collagen can be dialyzed against 0.01 N acetic acid, for example, prior to lyophilization to remove the sterilization solution and so that the isolated collagen is in a 0.01 N acetic acid solution.
  • the isolated collagen can be dialyzed against hydrochloric acid, for example, prior to lyophilization and can be lyophilized in hydrochloric acid and redissolved in hydrochloric acid, acetic acid, or water.
  • the resulting lyophilizate can be redissolved in any solution, but may be redissolved in an acidic solution or water.
  • the lyophilizate can be redissolved in, for example, acetic acid, hydrochloric acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid, at any of the above-described concentrations, or can be redissolved in water.
  • the lyophilizate is redissolved in 0.01 N acetic acid.
  • the redissolved lyophilizate can be subjected to varying conditions (e.g., pH, phosphate concentration, temperature, buffer composition, ionic strength, and composition and concentration of the isolated collagen components (dry weight/ml)) that result in polymerization to form engineered matrices for specific tissue graft applications.
  • varying conditions e.g., pH, phosphate concentration, temperature, buffer composition, ionic strength, and composition and concentration of the isolated collagen components (dry weight/ml)
  • the isolated collagen can be subjected to varying conditions for polymerization to form fibrils.
  • the conditions that can be varied include pH, phosphate concentration, temperature, buffer composition, ionic strength, the particular isolated collagen components (e.g., type I and type III collagen) and other ECM components present, and the concentration of the isolated collagen (dry weight/ml). These conditions result in polymerization of the isolated collagen to form engineered matrices with desired compositional, microstructural, and mechanical characteristics.
  • these compositional, microstructural, and mechanical characteristics can include fibril length, fibril diameter, number of fibril-fibril connections (e.g., cross-links), fibril density, fibril organization, matrix composition, 3-dimensional shape or form, and viscoelastic, tensile, shear, or compressive behavior (e.g., failure stress, failure strain, and modulus), permeability, degradation rate, swelling, hydration properties (e.g., rate and swelling), and in vivo tissue remodeling and bulking properties, rate of polymerization, lag time of polymerization, extent of polymerization, viscosity of interstitial fluid and desired in vitro and in vivo cell responses.
  • the collagen-based matrices described herein have desirable biocompatibility and in vitro and in vivo remodeling properties, among other desirable properties.
  • qualitative and quantitative microstructural characteristics of the engineered matrices can be determined by environmental or cryostage scanning electron microscopy, transmission electron microscopy, confocal microscopy, second harmonic generation multi-photon microscopy.
  • polymerization kinetics may be determined by spectrophotometry or time-lapse confocal reflection microscopy.
  • tensile, compressive and viscoelastic properties can be determined by rheometry or tensile testing.
  • a rat subcutaneous injection model can be used to determine remodeling properties. All of these methods are known in the art or are further described in the Examples or are described in Roeder et al., J. Biomech.
  • the isolated collagen is polymerized to form fibrils at a final concentration (dry weight/ml) of about 0.05 to about 5.0 mg/ml of the isolated collagen, or in another embodiment the final concentration is selected from the range of about 0.05 mg/ml to about 4.0 mg/ml, and in another embodiment the final concentration is selected from the range of about 0.05 mg/ml to about 3.0 mg/ml, and in another embodiment the final concentration is about 0.05, 0.1, 0.2, 0.3, 0.5, 1.0, 2.0, or 3.0 mg/ml.
  • the isolated collagen is polymerized at final concentrations (dry weight/ml) of about 5 to about 10 mg/ml, about 5 to about 30 mg/ml, about 5 to about 50 mg/ml, about 5 to about 100 mg/ml, about 20 to about 50 mg/ml, about 20 to about 60 mg/ml, or about 20 to about 100 mg/ml.
  • the polymerization reaction is conducted in a buffered solution using any biologically compatible buffer known to those skilled in the art.
  • the buffer may be selected from the group consisting of phosphate buffer saline (PBS), Tris(hydroxymethyl)aminomethane Hydrochloride (Tris-HCl), 3-(N-Morpholino) Propanesulfonic Acid (MOPS), piperazine-n,n′-bis(2-ethanesulfonic acid) (PIPES), [n-(2-Acetamido)]-2-Aminoethanesulfonic Acid (ACES), N-[2-hydroxyethyl]piperazine-N′-[2-ethanesulfonic acid] (HEPES), and 1,3-bis[tris(Hydroxymethyl)methylamino]propane (Bis Tris Propane).
  • PBS phosphate buffer saline
  • Tris-HCl Tris(hydroxymethyl)aminomethane Hydrochloride
  • the polymerization of the isolated collagen is conducted at a pH selected from the range of about 5.0 to about 11, and in one embodiment polymerization is conducted at a pH selected from the range of about 6.0 to about 9.0, and in one embodiment polymerization is conducted at a pH selected from the range of about 6.5 to about 8.5, and in another embodiment the polymerization of the isolated collagen is conducted at a pH selected from the range of about 7.0 to about 8.5, and in another embodiment the polymerization of the isolated collagen is conducted at a pH selected from the range of about 7.3 to about 7.4.
  • the ionic strength of the buffered solution is also regulated.
  • the ionic strength of the buffer used to polymerize the isolated collagen is selected from a range of about 0.05 to about 1.5 M, in another embodiment the ionic strength is selected from a range of about 0.10 to about 0.90 M, in another embodiment the ionic strength is selected from a range of about 0.14 to about 0.30 M and in another embodiment the ionic strength is selected from a range of about 0.14 to about 0.17 M.
  • the polymerization is conducted at temperatures selected from the range of about 0° C. to about 60° C. In other embodiments, the polymerization is conducted at temperatures above 20° C., and typically the polymerization is conducted at a temperature selected from the range of about 20° C. to about 40° C., and more typically the temperature is selected from the range of about 30° C. to about 40° C. In one illustrative embodiment the polymerization is conducted at about 37° C.
  • the phosphate concentration is varied.
  • the phosphate concentration is selected from a range of about 0.005 M to about 0.5 M.
  • the phosphate concentration is selected from a range of about 0.01 M to about 0.2 M.
  • the phosphate concentration is selected from a range of about 0.01 M to about 0.1 M.
  • the phosphate concentration is selected from a range of about 0.01 M to about 0.03 M.
  • the isolated collagen can be polymerized by, for example, extrusion into a desired buffer, including the buffers described above, or wet-spinning to form strands of isolated collagen.
  • the strands can be formed by extrusion through a needle and can be air-dried to form fibers or threads of various dimensions.
  • the syringe can be adapted with needles or tubing to control the dimensions (e.g., diameter) of the fibers or threads.
  • the extrusion process involves polymerization of the isolated collagen followed by extrusion into a bath containing water, a buffer, or an organic solvent (e.g., ethanol).
  • the extrusion process involves coextrusion of the isolated collagen with a polymerization buffer (e.g., the buffer such as Tris or phosphate buffers at various concentrations can be varied to control pH and ionic strength).
  • a polymerization buffer e.g., the buffer such as Tris or phosphate buffers at various concentrations can be varied to control pH and ionic strength.
  • the extrusion process involves extrusion of the isolated collagen into a polymerization bath (e.g., the buffer such as Tris or phosphate buffers at various concentrations can be varied to control pH and ionic strength).
  • the bath conditions affect polymerization time and properties of the fibers or threads, such as mechanical integrity of the fibers or threads, fiber dimensions, and the like.
  • the fibers can be air-dried to create materials suitable for use as sutures. Multiple fibers can be assembled (e.g., woven, braided to form matrix constructs).
  • the engineered matrices of the present invention can be combined, prior to, during, or after polymerization, with nutrients, including minerals, amino acids, sugars, peptides, proteins, vitamins (such as ascorbic acid), or glycoproteins that facilitate cellular proliferation, such as laminin and fibronectin, hyaluronic acid, or growth factors such as epidermal growth factor, platelet-derived growth factor, transforming growth factor beta, or fibroblast growth factor, and glucocorticoids such as dexamethasone.
  • fibrillogenesis inhibitors such as glycerol, glucose, or polyhydroxylated compounds can be added prior to or during polymerization.
  • cells can be added to the isolated collagen as the last step prior to the polymerization or after polymerization of the engineered matrix.
  • particulate extracellular matrix material can be added to the isolated collagen and can enhance bulking capacity.
  • cross-linking agents such as carbodiimides, aldehydes, lysl-oxidase, N-hydroxysuccinimide esters, imidoesters, hydrazides, and maleimides, and the like can be added before, during, or after polymerization.
  • the isolated collagen is derived from a collagen-containing source material and, in some embodiments, may contain glycoproteins, such as laminin and fibronectin, proteoglycans, such as serglycin, versican, decorin, and perlecan, and glycosaminoglycans.
  • the isolated collagen can be further purified or partially purified and the purified or partially purified composition can be used in accordance with the methods described herein or mixtures of partially purified or purified components can be used.
  • the term “purified” means the isolation of collagen in a form that is substantially free from other components (e.g., typically the total amount of other components present in the composition represents less than 5%, or more typically less than 0.1%, of total dry weight).
  • engineered matrices are provided.
  • the engineered matrices can be prepared according to the methods described herein, including any of the methods or conditions for polymerization described herein.
  • the engineered matrices comprise collagen fibrils.
  • collagen fibril refers to a quasi-crystalline, filamentous structure formed by the self-assembly of soluble collagen molecules.
  • the engineered matrices comprise collagen fibrils which may pack in a quarter-staggered pattern giving the fibril a characteristic striated appearance or banding pattern along its axis.
  • Collagen fibrils are distinct from the amorphous aggregates or precipitates of insoluble collagen that can be formed by dehydrating (e.g., lyophilizing) collagen suspensions to form porous network scaffolds.
  • the matrices are prepared from isolated collagen at collagen concentrations ranging from about 0.05 to about 5.0 mg/ml, about 1.0 mg/ml to about 3.0 mg/ml, about 0.05 mg/ml to about 10 mg/ml, about 0.05 to about 20 mg/ml, about 0.05 to about 30 mg/ml, about 0.05 to about 40 mg/ml, about 0.05 to about 50 mg/ml, about 0.05 to about 60 mg/ml, about 0.05 to about 80 mg/ml, about 5 mg/ml to 10 mg/ml, about 5 mg/ml to 20 mg/ml, about 5 mg/ml to about 40 mg/ml, about 5 mg/ml to 60 mg/ml, about 5 mg/ml to about 100 mg/ml, about 20 mg/ml to about 40 mg/ml, about 20 mg/ml to 60 mg/ml, or about 20 mg/ml to about 100 mg/ml.
  • the engineered matrices contain fibrils with specific characteristics, including, but not limited to, a fibril area fraction (defined as the percent area of the total area occupied by fibrils in a cross-sectional surface of the matrix; 2-dimensional) or a fibril volume fraction (the percent area of the total area occupied by fibrils in 3 dimensions) of about 0.1% to about 100%, about 0.5% to about 100%, about 0.5% to about 26%, about 1% to about 100%, about 1% to about 26%, about 1% to about 7%, about 1% to about 15%, of about 7% to about 26%, about 20% to about 30%, about 20% to about 50%, about 20% to about 70%, about 20% to about 100%, about 30% to about 50%, about 30% to about 70%, or about 30% to about 100%.
  • a fibril area fraction defined as the percent area of the total area occupied by fibrils in a cross-sectional surface of the matrix; 2-dimensional
  • a fibril volume fraction the percent area of the total area occupied by fibrils in 3 dimensions
  • the three-dimensional engineered matrices have a modulus (e.g., an elastic or linear modulus (defined by the slope of the linear region of the stress-strain curve obtained using conventional mechanical testing protocols; i.e., stiffness), a compressive modulus, or a shear storage modulus) of about 0.5 kPa to about 40 kPa, about 30 kPa to 100 kPa, about 30 kPa to about 1000 kPa, about 30 kPa to about 10000 kPa, about 30 kPa to about 70000 kPa, about 100 kPa to 1000 kPa, about 100 kPa to about 10000 kPa, or about 100 kPa to about 70000 kPa.
  • a modulus e.g., an elastic or linear modulus (defined by the slope of the linear region of the stress-strain curve obtained using conventional mechanical testing protocols; i.e., stiffness), a compressive modulus, or a shear
  • the engineered matrices have a fibril area fraction or a fibril volume fraction of about 0.5% to about 1%, about 0.5% to about 5%, 0.5% to about 10%, about 0.5% to about 50%, about 0.5% to about 100%, or of about 7% to about 26%. In another embodiment the engineered matrices have a fibril area fraction or a fibril volume fraction of about 7% to about 15% or about 16% to about 26%. In another embodiment the engineered matrices have a fibril area fraction or a fibril volume fraction of about 18.5% to about 25%.
  • the engineered matrices are formed from collagen at concentrations of about 3.2, 3.4, 3.6, 3.8, 4.0, 4.5 or 5.0 mg/ml of collagen, resulting in engineered matrices having a fibril area fraction of about 19%, 19.7%, 20.5%, 21.2%, 22%, 23.8% and 25.6%, respectively.
  • the engineered matrices have a modulus of about 0.5 to about 40 kPa. In accordance with another embodiment, the engineered matrices have a relatively low modulus of about 0.5 to about 24.0 kPa. In one other embodiment, the engineered matrices have a relatively high modulus of about 25 to about 40 kPa.
  • the polymerization reaction for engineered matrices can be conducted in a buffered solution using any biologically compatible buffer system known to those skilled in the art.
  • the buffer may be selected from the group consisting of phosphate buffer saline (PBS), Tris (hydroxymethyl)aminomethane Hydrochloride (Tris-HCl), 3-(N-Morpholino) Propanesulfonic Acid (MOPS), piperazine-n,n′-bis(2-ethanesulfonic acid) (PIPES), [n-(2-Acetamido)]-2-Aminoethanesulfonic Acid (ACES), N-[2-hydroxyethyl]piperazine-N′-[2-ethanesulfonic acid] (HEPES) and 1,3-bis[tris(Hydroxymethyl)methylamino]propane (Bis Tris Propane).
  • the buffer is PBS, Tris, or MO
  • the ratio of Na 2 HPO 4 and KH 2 PO 4 is varied.
  • ionic strength may be adjusted as an independent variable by varying the molarity of NaCl only.
  • the isolated collagen can be pipetted into a plate with wells and the isolated collagen can be allowed to polymerize under any of the conditions described above. For example, a humidified environment at 37° C. for approximately 30 minutes can be used. In an alternative embodiment, the isolated collagen is injected into a host and is polymerized in vivo.
  • cells can be added to the isolated collagen as the last step prior to the polymerization.
  • cells can be added after polymerization of the engineered matrix.
  • the engineered matrices comprising the cells can be subsequently injected or implanted in a host for use as a tissue graft.
  • the cells on or within the engineered matrix can be cultured in vitro, for a predetermined length of time, to increase the cell number or to induce desired remodeling prior to implantation or injection into a host.
  • the cells can be cultured in vitro, for a predetermined length of time, to increase cell number and the cells can be separated from the matrix and implanted or injected into the host in the absence of the engineered matrix.
  • the engineered matrices can include exogenous glucose and/or calcium chloride in the interstitial fluid of the matrices to promote cell growth. In one embodiment, about 1.0 mM to about 300 mM glucose and about 0.2 mM to about 4.0 mM CaCl 2 is included.
  • the isolated collagen comprises about 0.1 mg/ml to about 3 mg/ml total isolated collagen in about 0.05 to about 0.005N HCl, about 0.07M to about 0.28M NaCl, about 1.3 to about 4.5 mM KCl, about 4.0 to about 16 mM Na 2 HPO 4 , about 0.7 to about 3.0 mM KH 2 PO 4 , about 0.25 to about 1.0 mM MgCl 2 , and about 2.77 mM to about 166.5 mM glucose.
  • polymerization of the isolated collagen is induced by the addition of a neutralizing solution such as NaOH.
  • a NaOH solution can be added to a final concentration of 0.01N NaOH.
  • cells are then added and a calcium chloride solution is also added to bring the final concentration of CaCl 2 to about 0.4 mM to about 2.0 mM CaCl 2 .
  • the composition is then allowed to polymerize either in vitro or in vivo to form an engineered matrix comprising collagen fibrils with cells in and/or on the matrix.
  • a kit for preparing engineered matrices.
  • the kit comprises sterilized components that can be combined to form an engineered matrix comprising collagen fibrils.
  • cells may constitute a component of the kit.
  • the kit comprises an isolated collagen composition, and a polymerization composition.
  • the kit comprises separate vessels, each containing one of the following components: isolated collagen, a phosphate buffer solution, a glucose solution, a calcium chloride solution, and a basic neutralizing solution.
  • the isolated collagen is provided in a lyophilized form and the kit is further provided with a solution of acetic acid (or other dilute acid including for example, hydrochloric acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid) for resuspending the lyophilized isolated collagen.
  • acetic acid or other dilute acid including for example, hydrochloric acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid
  • the kit comprises a solution comprising isolated collagen, a phosphate buffer solution, a glucose solution, a calcium chloride solution, an acid solution, and a basic neutralizing solution.
  • the kit can include isolated collagen and a polymerization buffer.
  • the polymerization composition comprises a phosphate buffer that has a pH of about 7.2 to about 7.6 and the acid solution is an acetic acid solution comprising about 0.05N to about 0.005N acetic acid. In another embodiment, the acid solution is about 0.01N acetic acid.
  • the polymerization composition comprises a phosphate buffer that has a pH of about 7.2 to about 7.6 and the acid solution is a hydrochloric acid solution comprising about 0.05N to about 0.005N hydrochloric acid.
  • the acid solution is about 0.01N hydrochloric acid.
  • the glucose solution has a concentration selected from the range of about 0.2% to about 5% w/v glucose, or about 0.5% to about 3% w/v glucose, and in one embodiment the glucose solution is about 1% w/v glucose.
  • the CaCl 2 solution has a concentration selected from the range of about 2 mM to about 40.0 mM CaCl 2 or about 0.2 mM to about 4.0 mM CaCl 2 , or about 0.2 to about 2 mM CaCl 2 .
  • the kit is provided with a 10 ⁇ PBS buffer having a pH of about pH 7.4, and comprising about 1.37M NaCl, about 0.027M KCl, about 0.081M Na 2 HPO 4 , about 0.015M KH 2 PO 4 , about 5 mM MgCl 2 and about 1% w/v glucose.
  • kits are provided that comprise three-dimensional, preformed engineered matrices prepared according to any of the methods described herein and wherein the kits comprise any of the components described herein.
  • kits can further comprise instructional materials describing methods for mixing the kit reagents to prepare engineered matrices or describing methods for using preformed, three-dimensional engineered matrices.
  • the instructional materials can provide information regarding the final concentrations that give optimal microenvironmental conditions including fibril microstructure and mechanical properties for a particular cell type or for a particular desired result.
  • the lyophilized isolated collagen prepared by the methods described herein maintains its bioactivity (i.e., the capacity to polymerize and form fibrils in vitro or in vivo and to remodel tissue in vivo).
  • the isolated collagen can be used, for example, for making engineered matrices for specific tissue graft applications.
  • the lyophilized isolated collagen is useful commercially for the mass production of isolated collagen (i.e., the components can be lyophilized and stored without loss of bioactivity) for use in making engineered matrices. Lyophilized, isolated collagen that retains bioactivity and polymerization capacity is also useful for the concentration of isolated collagen for preparing engineered matrices that require concentration (i.e., higher concentrations) of collagen.
  • a collagen composition comprising collagen oligomers wherein the oligomers are isolated from a mammalian tissue enriched with or containing collagen oligomers.
  • a method for engineering matrices with enhanced polymerization characteristics comprises the steps of obtaining collagen oligomers, polymerizing the collagen oligomers, and forming the engineered matrices with enhanced polymerization characteristics.
  • the enhanced polymerization characteristics are selected from the group consisting of an enhanced rate of polymerization, a reduced lag time for polymerization, and an enhanced mechanical integrity.
  • the enhanced mechanical integrity is selected from the group consisting of strength and stiffness.
  • the polymerization characteristics are enhanced relative to type I isolated collagen obtained from Sigma-Aldrich as described in Example 2 and polymerized under the conditions described in Example 3.
  • the collagen oligomers polymerize with a maximal T 1 ⁇ 2 of 3 minutes, 2.5 minutes, 2.0 minutes, 1.5 minutes, or 1 minute.
  • the collagen-containing source material enriched with collagen oligomers is a tissue naturally enriched with collagen oligomers.
  • the collagen-containing source material enriched with collagen oligomers is a diseased tissue or a genetically-modified tissue where the tissue is enriched with collagen oligomers.
  • the collagen-containing source material enriched with collagen oligomers is a mechanically-modified tissue or an electrically-modified tissue.
  • the collagen-containing source material is mechanically modified cultured cells or electrically-modified cultured cells.
  • the collagen oligomers are obtained by isolating and then chemically cross-linking collagen.
  • Extracellular matrix materials can be utilized as tissue graft compositions for remodeling tissues in vivo or for in vitro applications.
  • the methods of collagen isolation and the collagen compositions described herein allow for the controlled alteration of the microstructural and subsequent mechanical properties of a resulting engineered matrix for such uses as graft compositions for inducing the repair of damaged or diseased tissues in vivo, and for inducing the proliferation or other fundamental behaviors of cells in vitro.
  • Systematic variation of engineered matrix design features using the methods of polymerization described herein provides control of in vivo (e.g., cell infiltration, vascularization, rate of cell proliferation, differentiation, morphogenesis, remodeling, degradation, and contractility) and in vitro (e.g., cell morphology, migration, proliferation, differentiation, morphogenesis, and contractility) responses.
  • in vivo e.g., cell infiltration, vascularization, rate of cell proliferation, differentiation, morphogenesis, remodeling, degradation, and contractility
  • in vitro e.g., cell morphology, migration, proliferation, differentiation, morphogenesis, and contractility
  • a graft composition comprising an engineered matrix comprising collagen oligomers.
  • the graft composition has a predetermined percentage of collagen oligomers based on total isolated collagen added to make the engineered matrix.
  • the predetermined percentage of collagen oligomers can be about 0.5% to about 100%, about 1% to about 100%, about 2% about 100%, about 3% to about 100%, about 5% to about 100%, about 10% to about 100%, about 15% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 90% to about 100%, about 95% to about 100%, or about 100%.
  • the collagen oligomers are obtained from a collagen-containing source material enriched with collagen oligomers (e.g., pig skin).
  • the collagen-containing source material enriched with collagen oligomers is a natural tissue, a diseased tissue or a genetically-modified tissue where the tissue is enriched with collagen oligomers.
  • the graft composition comprises collagen oligomers obtained by isolating and then chemically cross-linking collagen.
  • the polymerization characteristics of the engineered matrix used to make the graft composition are enhanced relative to type I isolated collagen obtained from Sigma-Aldrich as described in Example 2 and polymerized under the conditions described in Example 3.
  • the collagen oligomers that polymerize to form the engineered matrix polymerize with a maximal T 1 ⁇ 2 of 3 minutes, 2.5 minutes, 2.0 minutes, 1.5 minutes, or 1 minute.
  • isolated collagen enriched with oligomers can be prepared by isolating collagen from a natural tissue (e.g., pig skin) according to the methods described herein.
  • the collagen oligomers can be isolated by a method such as the method described in Example 10.
  • oligomer-monomer separation can be achieved using methods that vary polymerization reaction parameters including temperature, pH, and ionic strength. Any other method known to the skilled artisan to be useful for separating collagen oligomers and monomers can be used.
  • a kit for preparing engineered matrices.
  • the kit comprises sterilized components that can be combined to form an engineered matrix comprising collagen fibrils.
  • the sterilized components include isolated collagen oligomers.
  • cells may constitute a component of the kit.
  • the kit comprises an isolated collagen oligomer composition, and a polymerization composition.
  • the kit comprises separate vessels, each containing one of the following components: isolated collagen oligomers, a phosphate buffer solution, a glucose solution, a calcium chloride solution, and a basic neutralizing solution.
  • the kit comprises separate vessels, each containing one of the following components: isolated collagen oligomers, isolated collagen monomers, a phosphate buffer solution, a glucose solution, a calcium chloride solution, and a basic neutralizing solution.
  • isolated collagen is provided in a lyophilized form and the kit is further provided with a solution of acetic acid (or other dilute acid including for example, hydrochloric acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid) for resuspending the lyophilized isolated collagen oligomers or monomers.
  • acetic acid or other dilute acid including for example, hydrochloric acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid
  • the kit comprises a solution comprising isolated collagen oligomers and/or monomers, a phosphate buffer solution, a glucose solution, a calcium chloride solution, an acid solution, and a basic neutralizing solution.
  • the kit can comprise isolated collagen and a polymerization buffer.
  • the polymerization composition comprises a phosphate buffer that has a pH of about 7.2 to about 7.6 and the acid solution is an acetic acid solution comprising about 0.05N to about 0.005N acetic acid. In another embodiment, the acid solution is about 0.01N acetic acid.
  • the polymerization composition comprises a phosphate buffer that has a pH of about 7.2 to about 7.6 and the acid solution is a hydrochloric acid solution comprising about 0.05N to about 0.005N hydrochloric acid. In another embodiment, the acid solution is about 0.01N hydrochloric acid. In one embodiment, the glucose solution has a concentration selected from the range of about 0.2% to about 5% w/v glucose, or about 0.5% to about 3% w/v glucose, and in one embodiment the glucose solution is about 1% w/v glucose.
  • the CaCl 2 solution has a concentration selected from the range of about 2 mM to about 40.0 mM CaCl 2 or about 0.2 mM to about 4.0 mM CaCl 2 , or about 0.2 to about 2 mM CaCl 2 .
  • the kit is provided with a 10 ⁇ PBS buffer having a pH of about pH 7.4, and comprising about 1.37M NaCl, about 0.027M KCl, about 0.081M Na 2 HPO 4 , about 0.015M KH 2 PO 4 , about 5 mM MgCl 2 and about 1% w/v glucose.
  • kits comprise three-dimensional, preformed engineered matrices prepared according to any of the methods described herein, including engineered matrices formed from isolated collagen oligomers alone, or isolated collagen oligomers and monomers in predetermined percentages of total collagen used to form the matrix, and wherein the kits comprise any of the components described herein.
  • kits can further comprise instructional materials describing methods for mixing the kit reagents to prepare engineered matrices or describing methods for using preformed, three-dimensional engineered matrices.
  • the instructional materials can provide information regarding the final concentrations that give optimal microenvironmental conditions including fibril microstructure and mechanical properties for a particular cell type or for a particular desired result.
  • tissue was harvested from pig immediately following euthanasia and was washed thoroughly with cold water. The skin was stretched out and pinned to a board and stored at 4° C. The hair was removed with clippers. The dermal layer of the tissue was isolated by separating and removing the upper epidermal layer and the lower loose fatty connective layers. This removal was readily achieved by scraping the tissue with a knife or straight razor. The tissue was maintained at 4° C.
  • the resulting dermal layer tissue was washed in water and then cut into small pieces (approximately 1 cm2) and was frozen and stored at ⁇ 80° C.
  • the frozen skin pieces were pulverized under liquid nitrogen using an industrial blender or cryogenic grinder. Soluble proteins were removed by extracting the pig skin powder (0.125 g/ml) with 0.5M sodium acetate overnight at 4° C. The resulting mixture was then centrifuged at 2000 rpm (700 ⁇ g) at 4° C. for 1 hour. The supernatant was discarded and the extraction procedure repeated three additional times.
  • the resulting pellet was then suspended (0.25 g/ml) in cold MilliQ water and then centrifuged at 2000 rpm (700 ⁇ g) at 4° C. for 1 hour. The pellet was then washed with water two additional times. Collagen extraction was then performed by suspending the pellet (0.125 g/ml) in 0.075M sodium citrate. The extraction was allowed to proceed for 15-18 hours at 4° C.
  • the resulting mixture was centrifuged at 2000 rpm (700 ⁇ g) at 4° C. for 1 hour. The supernatant was retained and stored at 4° C. The pellet was re-extracted with 0.075M sodium citrate. The extraction process was repeated such that the tissue was extracted a total of three times. The resulting supernatants were then combined and centrifuged at 9750 rpm (17,000 ⁇ g) at 4° C. for 1 hour to clarify the solution. The supernatant was retained and the pellet discarded.
  • Collagen was then precipitated from the supernatant by dialyzing (MWCO 12-14,000) extensively against 0.02 M disodium hydrogen phosphate at 4° C. The resulting suspension was then centrifuged at 2000 rpm at 4° C. for 1 hour and the pellet retained. The pellet was then resuspended and rinsed in cold MilliQ water. The suspension was centrifuged at 2000 rpm at 4° C. for 1 hour. The water rinse procedure was repeated two additional times.
  • the resulting collagen pellet was dissolved in 0.1M acetic acid and then lyophilized.
  • the lyophilized material was stored within a dessicator at 4° C. for use in engineering ECMs.
  • Interrupted gel electrophoresis was performed to compare the protein composition of the collagen preparations made in accordance with the methods described herein to commercially available type I isolated collagen obtained from Sigma-Aldrich, St. Louis, Mo. (Sigma Cat. No. C3511) and pepsin-solubilized collagen from INAMED (Fremont, Calif.) sold under the product name PureColTM. Interrupted gel electrophoresis was performed according to Sykes, et al. The estimation of two collagens from human dermis by interrupted gel electrophoresis. Biochem. Biophys. Res. Commun. 72:1472-1480 (1976), incorporated herein by reference.
  • SDS-PAGE Sodium dodecylsulfate polyacryamide gel electrophoresis
  • the evaluation by SDS-PAGE of the protein composition of the collagen preparations made in accordance with the methods described herein indicates that the protocol yielded a pure type I collagen preparation (see below) with minimal to no contaminating non-collagenous proteins (see lanes 2-4 of the gel shown in FIG. 1 ).
  • the gel shown in FIG. 1 indicates the band pattern of the ⁇ 1, ⁇ 2, ⁇ 11, and ⁇ 12 chains of collagen. No significant glycosaminoglycan content was identified using a standard alcian blue assay (Bjornsson, S. Simultaneous preparation and quantitation of proteoglycans by precipitation with alcian blue. Anal Biochem. 210:282-291 (1993), incorporated herein by reference).
  • Cyanogen bromide (CNBr) peptide analysis was used to confirm the presence of type I collagen in the pig skin collagen preparation (lanes 2-4), made according to the methods described herein.
  • CNBr peptide analysis was performed according to the method of Miller, et al. Identification of three genetically distinct collagens by cyanogen bromide cleavage of insoluble skin and cartilage collagen. Biochem. Biophys. Res. Commun. 42:1024-1029 (1971). Standard SDS-PAGE within 12% gels was then performed to compare the CNBr-derived peptides from the various collagen preparations (see FIG. 6 ). The pattern shown in FIG.
  • pig skin collagen preparation made in accordance with the methods described herein, has a number of bands which are similar to Sigma collagen and INAMED collagen. However, there are also bands specific to the pig skin collagen composition. The presence of type I collagen was confirmed in the preparation described herein, and the preparation described herein does not have significant amounts of type V collagen or type III collagen (confirmed using interrupted gel electrophoresis). Types III and V collagen are the other two major types of collagen found in skin.
  • the pig skin type I collagen preparation (lanes 2-4), made according to the methods described herein, yielded a distinct collagen banding pattern compared to the commercially available collagen preparations, namely Sigma Type I collagen (acid solubilized; lane 5) and INAMED collagen (acid/pepsin solubilized; lane 6).
  • the collagen preparations made in accordance with the methods described herein were observed.
  • band designated “Unknown” a unique high molecular weight protein band was identified within the pig skin collagen preparation made in accordance with the methods described herein. The band was not observed in the INAMED or Sigma commercially available collagen preparations.
  • the unknown protein band was also isolated, digested with CNBr, and a banding pattern indicative of collagen alpha chains was found.
  • the polymerization kinetics of the pig skin collagen preparation made in accordance with the methods described herein were also compared to commercially available Sigma Type I collagen.
  • a spectrophotometric turbidity analysis assay for determining polymerization kinetics of collagen well-known to those skilled in the art was used.
  • the assay was performed in accordance with Comper, et al. Characterization of nuclei in in vitro collagen fibril formation. Biopolymers 16:2133-2142 (1977) and Brightman, et al. Time-lapse confocal reflection microscopy of collagen fibrillogenesis and extracellular matrix assembly in vitro. Biopolymers 54:222-234 (2000), each incorporated herein by reference.
  • the time-course of polymerization was monitored in a Lambda 35 UV-VIS spectrophotometer (Perkin-Elmer) equipped with a temperature-controlled, 8-position cell changer as described previously by Brightman et al., 2000.
  • FIG. 2 shows the time-dependent changes in absorbance as recorded at a wavelength of 405 nm as each of the collagen preparations (each at 2 mg/ml collagen concentration as determined using sirius red analysis) underwent polymerization or fibrillogenesis (i.e., soluble state to an insoluble fibrillar state).
  • the sirius red assay was performed in accordance with Marotta, et al. Sensitive spectrophotometric method for the quantitative estimation of collagen. Anal. Biochem. 150:86-90 (1985), incorporated herein by reference.
  • the pig skin collagen preparation made in accordance with the methods described herein showed a decreased lag time, increased polymerization rate, and increased magnitude of change in A405 readings.
  • the pig skin collagen preparation made in accordance with the methods described herein exhibited a much decreased lag time and increased polymerization rate in comparison to commercially available Sigma Type I collagen.
  • the collagen fibril microstructure of a 3D ECM engineered from pig skin collagen, prepared in accordance with the methods described herein, and from commercially available Sigma Type I collagen were determined and compared using confocal reflection microscopy. Each collagen composition was polymerized at 1.5 mg/ml. Confocal Reflection Microscopy was performed according to Brightman, et al. Time-lapse confocal reflection microscopy of collagen fibrillogenesis and extracellular matrix assembly in vitro. Biopolymers 54:222-234 (2000) and Voytik-Harbin, et al. Three-dimensional imaging of extracellular matrix and extracellular matrix-cell interactions. Methods in Cell Biology 63:583-597 (2001), each incorporated herein by reference.
  • solutions of collagen were polymerized in a Lab-Tek chambered coverglass and imaged using a BioRad Radiance 2100 MP Rainbow confocal/multiphoton microscope using a 60 ⁇ 1.4 NA oil immersion lens (or an Olympus Fluoview FV 1000 confocal microscope was used).
  • Optical settings were established and optimized for matrices after polymerization was complete. Samples were illuminated with 488 nm laser light and the reflected light detected with a photomultiplier tube (PMT) using a blue reflection filter.
  • PMT photomultiplier tube
  • the fibril microstructure produced by the pig skin collagen, prepared in accordance with the methods described herein, showed an increase in fibril density and, surprisingly, an apparent increase in fibril-to-fibril association (e.g., cross-linked and/or fibril aggregates; see FIGS. 3A (pig skin collagen preparation)) and 3 B (commercially available Sigma Type I collagen)).
  • the collagen fibril microstructure of a 3D ECM engineered from pig skin collagen, prepared in accordance with the methods described herein, and from commercially available Sigma Type I collagen were determined and compared using scanning electron microscopy. Each collagen composition was polymerized at 1.0 mg/ml.
  • FIGS. 4A and B show pig skin collagen, prepared in accordance with the methods described herein, and FIGS. 4C and D show commercially available Sigma Type I collagen.
  • Sigma collagen provided a less dense fibril architecture compared to the pig skin collagen. Scanning electron microscopy was performed according to Voytik-Harbin, et al.
  • Small intestinal submucosa A tissue-derived extracellular matrix that promotes tissue-specific growth and differentiation of cells in vitro. Tissue Engineering, 4:157-174 (1998), incorporated herein by reference. Note that scanning electron microscopy analysis involves fixing and critical point drying and the required processing of the specimens is known to lead to microstructural artifacts. Therefore, this technique alone is not relied on to determine the 3D microstructure of engineered ECMs.
  • FIG. 5 shows the mechanical behavior of a 3D ECM engineered from pig skin collagen (polymerized at 2 mg/ml), prepared in accordance with the methods described herein, and from commercially available Sigma Type I collagen (polymerized at 3 mg/ml). Mechanical behavior was compared using unconfined compression. The specimen was subjected to unconfined compression at a rate of 10 or 20 micrometers/sec using an AR-2000 rheometer (TA Instruments, New Castle, Del.). As shown in FIG. 5 , pig skin collagen, prepared in accordance with the methods described herein, surprisingly exhibited as much as 10-20 ⁇ greater mechanical integrity as commercially available Sigma Type I collagen.
  • collagen was polymerized by using a 10 ⁇ PBS, pH 7.4 solution consisting of 1.37M NaCl, 0.027M KCl, 0.081M Na 2 HPO 4 , 0.015M KH 2 PO 4 , 5 mM MgCl 2 , and 1% w/v glucose.
  • a mixture was made of 1 ml of solubilized collagen in 0.01N HCl, 150 ⁇ l 10 ⁇ PBS, pH 7.4, 150 ⁇ l 0.1N NaOH, 100 ⁇ l 13.57 mM CaCl 2 , and 100 ⁇ l 0.01 N HCl.
  • the composition was mixed well after each component was added. Polymerization was allowed to proceed at 37° C.
  • the shear storage modulus for ECMs prepared from pig skin collagen is 10 to 40 times greater than that obtained for ECMs prepared from commercially available Sigma collagen.
  • the pig skin collagen preparation made in accordance with the methods described herein, has been seeded with a number of different cell types, including human mesenchymal stem cells (Clonetics) and a mesenchymal stem cell line derived from the bone marrow of mice (D1, American Type Culture Collection (ATCC)).
  • human mesenchymal stem cells Clonetics
  • mesenchymal stem cell line derived from the bone marrow of mice
  • ATCC American Type Culture Collection
  • engineered ECMs were prepared using pig skin collagen, prepared according to the methods described herein, or commercially available Sigma collagen preparations at two concentrations, specifically 0.5 mg/ml and 3 mg/ml. These formulations were used to entrap and culture mouse mesenchymal stem cells within a 3D format. Results showed that engineered ECMs of varied microstructures and mechanical properties (stiffness) could be prepared from pig skin collagen. In turn, mouse mesenchymal stem cells cultured within pig skin ECMs of relatively low stiffness (0.5 mg/ml) showed preferential differentiation down the adipogenic pathway ( FIG. 8A ) while those prepared with relatively high stiffness (3 mg/ml) supported enhanced osteogenic differentiation ( FIG. 8B ).
  • ECMs prepared with commercially available Sigma collagen those prepared from pig skin supported enhanced osteogenesis.
  • the differences in the two preparations to support osteogenic differentiation of multi-potential stem cells can be attributed to differences in ECM microstructure and mechanical properties.
  • Engineered ECMs of increased mechanical integrity are much more readily obtained using the pig skin collagen preparation, made according to the methods described herein.
  • the cells in FIG. 1 are much more readily obtained using the pig skin collagen preparation, made according to the methods described herein.
  • FIG. 8 A were cultured for periods of time up to 3 weeks in DMEM supplemented with 10% fetal bovine serum, 0.5 mM isobutyl-methylxanthine, 1 ⁇ M dexamethasone, 10 ⁇ M insulin, 200 ⁇ M indomethacin, 0.3 mg/ml glutamine, 100 U/ml penicillin, and 100 ⁇ g/ml streptomycin.
  • the cells in FIG. 8 B were cultured for periods of time up to 3 weeks in DMEM supplemented with 10% fetal bovine serum, 0.3 mg/ml glutamine, 100 U/ml penicillin, and 100 ⁇ g/ml streptomycin.
  • Type I collagen isolated from pig skin was solubilized in acetic acid (0.005 M) to achieve a desired collagen concentration (4 mg/ml).
  • An equal volume of 2 ⁇ solubilization buffer was added (0.06 M phosphate, 0.2 M NaCl, and 1.2 M glycerol, pH 7.0) with mixing.
  • the solution was left to stand at 30° C. for 7 days. At desired times, the solution was then centrifuged at 10,000 rpm and 4° C. for 10 minutes and decanted and the supernatant was retained. The pellet was then resolubilized in 0.1 M acetic acid and the resolubilized pellet and the supernatant were dialyzed against 0.01 M acetic acid and lyophilized to dryness.
  • Confocal reflection (CRM) microscopy was used to collect high resolution images of ECMs (without cells). Fibril density and fibril diameters were measured from the images to quantify the effect of HA concentration on the collagen fibrillar microstructure. Confocal imaging was performed on a Olympus Fluoview FV1000 confocal system adapted to a IX81 inverted microscope (Olympus, Tokyo, Japan). For the fibril density, confocal image stacks were collected from random locations within independent ECMs for each treatment. The fibril density for each image was calculated in Matlab (Mathworks, Natick, Mass.) as the ratio of fibril volume (voxels containing fibrils) to total volume after binarizing (thresholding) the images.
  • Fibril diameters were measured from the 2D projections of the confocal images in Imaris 5.0 (Bitplane Inc., Saint Paul, Minn.). Each fibril diameter represents the average of 5 diameters measured along the major axis of an individual fibril.
  • the geometry was spun lightly and the geometry was lowered to a 725 ⁇ m gap position (this position correlates to 1 ml). The spinning motion helped to insure that the sample was evenly distributed under the geometry. Once the geometry was at the proper gap, it was verified that the collagen solution extended to the edge of the plate, but not past it. The vapor trap was placed on the plate, and the normal force was zeroed. A reading was then taken.
  • FIG. 9 shows the time-dependent glycerol-based separation of pig skin collagen type I oligomers and monomers.
  • the supernatant and pellet represent the monomer-rich and oligomer-rich fractions, respectively.
  • the results in FIG. 9 show that the pellet is enriched in the higher molecular weight oligomeric collagen molecules.
  • FIG. 10A shows glycerol-based separation of pig skin collagen type I oligomers and monomers.
  • Pig skin collagen was polymerized in the presence of glycerol for 7 days as described in Example 10. The mixture was centrifuged and the supernatant and pellet retained, dialyzed against dilute acetic acid, and lyophilized to dryness. The supernatant and pellet represented monomer- and oligomer-rich collagen compositions, respectively, as determined by SDS-PAGE (4%).
  • FIG. 10A shows the SDS-PAGE gel of the samples and
  • FIG. 10B shows the corresponding densitometry readings. With reference to the labeling of bands shown in FIG.
  • the densitometry readings show a comparison of the oligomer-rich and monomer-rich fraction densitometry readings for individual bands on the SDS-PAGE gel relative to densitometry readings for alpha 1.
  • the leftmost bar for each group of 3 bars is the initial (before glycerol separation) densitometry reading for each band relative to alpha 1 in the initial isolated collagen preparation
  • the middle bar for each group of 3 bars is the desitometry reading for each band in the oligomer-rich fraction after glycerol-based separation relative to alpha 1
  • the rightmost bar for each group of 3 bars is the desitometry reading for each band in the monomer-rich fraction after glycerol-based separation relative to alpha 1.
  • the results show that for known oligomers (e.g., ⁇ 12), the oligomers are found mainly in the pellet after glycerol-based separation. SDS-PAGE analysis was performed as described in Example 2.
  • FIG. 11 shows SDS-PAGE (12%) analysis of CNBr peptide maps of collagen preps in which oligomer:monomer content is varied. Upon CNBr digestion, collagen preps yielded distinct peptide fingerprints indicating distinct molecular compositions and cross-linked moieties. SDS-PAGE analysis and CNBr peptide analysis were performed as described in Example 3.
  • FIGS. 12 A-B show the oligomer:monomer content of pig skin collagen preparation affects polymerization (self-assembly) kinetics.
  • An increase in oligomer content decreases lag time ( FIG. 12 B) and increases the rate of polymerization ( FIG. 12 A).
  • FIG. 12A shows A 405 vs time.
  • the uppermost line is the control (0.7 mg/ml collagen) which is the resuspended collagen pellet after collagen isolation, but prior to glycerol separation.
  • the lowest line is 100% monomer-rich collagen.
  • the middle lines from top to bottom are 1.) 5.4% monomer-rich collagen and 94.6% oligomer-rich collagen, 2.) 100% oligomer-rich collagen, and 3.) 55.4% monomer rich+44.6% oligomer-rich collagen.
  • FIG. 13 shows the results of an assay done to compare mechanical behavior of 3D engineered matrices prepared with pig skin collagen preps of varied monomer:oligomer content.
  • the shear storage modulus (G′) provides a measure of the stiffness of the solid (elastic) components.
  • the results in FIG. 13 show that the shear storage modulus (G′; stiffness) of matrices increases with oligomer content.
  • the top line is the control.
  • the bottom line is 100% monomer-rich collagen.
  • the middle lines are (from top to bottom) 5.4% monomer-rich collagen and 94.6% oligomer-rich collagen, 100% oligomer-rich collagen, and 55.4% monomer rich+44.6% oligomer-rich collagen.
  • FIG. 14 shows the comparison of mechanical behavior of 3D engineered matrices prepared with pig skin collagen preps of varied monomer:oligomer content. An increase in delta as a function of strain as observed with the monomer-rich preparation (uppermost line) indicates more fluid-like (viscous) behavior.
  • FIG. 15 shows a comparison of mechanical behavior of 3D engineered matrices prepared with pig skin collagen preps of varied monomer:oligomer content. Matrices produced with collagen preps consisting of higher oligomer content show increased compressive stiffness. The bottom line is monomer-rich collagen. The next line towards the top is the control. The top three lines are, from top to bottom, 100% oligomer-rich collagen, 5.4% monomer-rich collagen and 94.6% oligomer-rich collagen, and 55.4% monomer rich+44.6% oligomer-rich collagen.
  • FIG. 16 shows the results of an assay done to determine the compressive stiffness values as a function of the oligomer:monomer content of the pig skin collagen preparation. The results show that stiffness increases with increasing oligomer content.
  • FIG. 17 shows the effects of oligomer content on the 3D collagen fibril microstructure of engineered matrices.
  • the matrices were prepared with the percentages of oligomers and monomers shown.
  • the oligomer-rich and monomer-rich preparations were 100% oligomer-rich and 100% monomer-rich, respectively. Confocal microscopy was performed as described in Example 4.
  • FIG. 18 shows the effects of oligomer content on the 3D collagen fibril microstructure of engineered matrices.
  • the matrices were prepared with the percentages of oligomers and monomers shown.
  • the oligomer-rich and monomer-rich preparations were 100% oligomer-rich and 100% monomer-rich, respectively. Confocal microscopy was performed as described in Example 4.
  • FIG. 19 shows the compositional analysis of different collagen preparations as determined using SDS-PAGE (4%). The results in FIG. 19 show that the pig skin collagen preparation uniquely contains collagen oligomers that run intermediate between commonly identified dimeric ( ⁇ ) and trimeric ( ⁇ ) forms of collagen. Upon pepsin treatment of pig skin collagen, the content of unique collagen oligomers as well as other higher molecular weight collagen species was significantly reduced.
  • FIG. 20 shows the kinetics of polymerization for the different collagen preparations as determined using the spectrophotometric-based turbidity assay described in Example 3. Results show that the polymerization kinetics of collagen is highly dependent upon the collagen preparation. More specifically, polymerization of pig skin collagen shows a decreased lag phase and an increased rate of polymerization (slope of growth phase). Pig skin collagen (0.25 mg/ml) and pig skin collagen (1.0 mg/ml) are the lower and upper left-most lines, respectively. Sigma collagen (0.25 mg/ml) and Sigma collagen (1.0 mg/ml) are the lowest line and the line where the third highest absorbance was achieved, respectively. PureCol (0.25 mg/ml) and PureCol (1.0 mg/ml) are the line with the greatest lag time and the line where the highest absorbance was achieved, respectively.
  • FIG. 21 shows a comparison of final absorbance (405 nm) values for different collagen preparations as determined from a spectrophometric based turbidity assay as described in Example 3. Results represent average of the last 10 absorbance values.
  • FIG. 22 shows the collagen fibril microstructure of 3D engineered ECMs prepared with different purified type I collagen sources. Each collagen preparation was polymerized at two different concentrations (0.5 mg/ml and 2 mg/ml) using the same reaction conditions. Images represent 2D projections of 3D image stacks as collected using confocal reflection microscopy as described in Example 4. The results show that the collagen fibril microstructure of 3D engineered ECMs is highly dependent upon the collagen preparation.
  • FIG. 23 shows the comparison of mechanical behavior of 3D engineered ECMs prepared with different collagen preparations and tested in oscillatory shear.
  • the storage modulus (G′) provides a measure of the stiffness of the solid (elastic) components. Results show that the stiffness of matrices prepared with pig skin collagen are significantly greater than those obtained with the commercial collagen preparations (PureCol and Sigma).
  • FIGS. 24A-B show a comparison of mechanical behaviors of engineered matrices prepared with different collagen sources.
  • FIG. 24 A shows microstructural analysis of engineered ECMs using confocal reflection microscopy (fibril density vs. collagen concentration).
  • FIG. 24 B shows microstructural analysis of engineered ECMs using confocal reflection microscopy (average fibril diameter vs. collagen concentration).
  • Collagen stiffness (G′ vs. fibril density) and rheometric analysis of mechanical properties (G′ (Pa) vs. collagen concentration) were also performed. Confocal microscopy was as described in Example 4.
  • Table 1 shows the effect of oligomer content on polymerization efficiency. Polymerization was assayed as described in Example 4. The collagen that remained in solution as polymerized versus non-polymerized collagen was determined based on a Sirius Red assay. The results in table 1 show that oligomer-rich collagen polymerizes more efficiently than monomer-rich collagen.
  • Table 1 shows the t 1 ⁇ 2's for polymerization for different collagen preparations. Polymerization was assayed as described in Example 4. The results in Table 2 show that oligomer-rich pig skin collagen polymerizes with a t 1 ⁇ 2 that is much lower than Sigma collagen or PureCol. A t 1 ⁇ 2 is the half-time required to achieve maximal absorbance.

Abstract

Collagen compositions, methods for preparing those collagen compositions, and graft compositions formed from those collagen compositions are provided. In particular, methods of isolating collagen that exhibits an enhanced rate of polymerization and enhanced microstructural and mechanical properties upon polymerization, such collagen compositions, and graft compositions formed from such collagen compositions are provided.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Application Ser. No. 60/846,207, filed on Sep. 21, 2006, incorporated by reference herein in its entirety.
  • FIELD OF THE INVENTION
  • This invention relates to collagen compositions, methods for preparing those collagen compositions, and graft compositions formed from those collagen compositions. More particularly, the invention relates to methods of isolating collagen that exhibits an enhanced rate of polymerization and enhanced micro-structural and mechanical properties upon polymerization and to such collagen compositions and graft compositions formed from such collagen compositions.
  • BACKGROUND AND SUMMARY
  • The extracellular matrix (ECM) as it occurs in vivo plays a crucial role in the organization, homeostasis, and function of tissues and organs. Continuous communication between cells and their surrounding ECM environment orchestrates critical processes such as the acquisition and maintenance of differentiated phenotypes during embryogenesis, the development of form (morphogenesis), angiogenesis, wound healing, and even tumor metastasis. Both biochemical and biophysical signals from the ECM modulate fundamental cellular activities including adhesion, migration, proliferation, differential gene expression, and programmed cell death. The realization of the significance of the ECM to the organization, homeostasis, and function of tissues and organs has led to a renewed interest in characterizing ECM constituents and the relationship of these constituents to the functioning of the ECM.
  • ECMs, including various basement membrane tissues and other extracellular matrix tissues obtained from natural sources and matrices formed from isolated ECM components, can be utilized as tissue graft compositions for remodeling tissues in vivo or for in vitro applications. Complex scaffolds representing combinations of isolated extracellular matrix components in a natural or processed form are commercially available and can be used as tissue graft compositions (e.g., Human Extracellular Matrix (Becton Dickinson) and MATRIGEL®). Basement membrane tissues and other extracellular matrix tissues, such as tissue material derived from submucosal tissues, harvested from warm-blooded vertebrates have also shown great promise as unique graft materials for inducing the repair of damaged or diseased tissues in vivo, and for inducing the proliferation of cells in vitro.
  • In accordance with the invention, purified collagen can be used to produce an engineered ECM material prepared under conditions that regulate the polymerization of collagen in a controlled manner. This result is more difficult to achieve with existing intact or processed ECMs from natural sources and with ECMs and collagen preparations from commercial sources.
  • In the literature, there are known methods for isolating collagen from a variety of tissues, e.g., placenta, bladder, animal tails, and skin, and using the isolated material to reconstitute collagenous matrices. These collagenous matrices may have applications as graft materials for inducing the repair of damaged or diseased tissues in vivo, and for inducing the proliferation and other fundamental behavior of cells in vitro. The molecular forces that orchestrate the self assembly of soluble, monomeric collagen into higher ordered structures are weak so their assembly can easily turn into an unstructured aggregation of misfolded proteins. As reported herein, modifying the conditions used to isolate collagen results in collagen preparations with properties that enhance the rate at which the collagen polymerizes and that enhance the microstructural and mechanical properties of the collagen upon polymerization (e.g., the mechanical integrity of the engineered ECM that is formed upon collagen polymerization). The methods of collagen isolation and the collagen compositions described herein allow for the controlled alteration of the microstructural and subsequent mechanical properties of a resulting engineered ECM for such uses as graft compositions for inducing the repair of damaged or diseased tissues in vivo, and for inducing the proliferation and other fundamental behaviors of cells in vitro.
  • In one embodiment, a method for isolating collagen is provided. The method comprises the steps of obtaining a collagen-containing source material, comminuting the source material, mixing the comminuted source material with an extraction solution, extracting the comminuted source material to form a soluble fraction and an insoluble fraction, obtaining the insoluble fraction, extracting the collagen from the insoluble fraction to form a soluble collagen fraction, precipitating the collagen from the soluble collagen fraction, and resuspending the precipitate in an aqueous solution wherein the aqueous solution used to resuspend the collagen precipitate is an acidic solution. An isolated collagen composition prepared by this method is also provided.
  • In various embodiments of the embodiment described in the preceding paragraph: 1) the collagen-containing source material is selected from the group consisting of placental tissue, bladder tissue, intestinal tissue, alimentary tract tissue, ovarian tissue, pericardial tissue, animal tail tissue, liver tissue, skin tissue, and any other suitable collagen-containing source material, 2) the collagen-containing source material is selected from the group consisting of bladder tissue and skin tissue, 3) the collagen-containing source material is porcine skin tissue, 4) the source material is frozen in liquid nitrogen prior to the comminuting step, 5) the source material is frozen in liquid nitrogen during the comminuting step, 6) the source material is frozen in liquid nitrogen prior to and during the comminuting step, 7) the source material is frozen at a temperature of −20° C. or below prior to or during the comminuting step, 8) the source material is frozen at a temperature of −40° C. or below prior to or during the comminuting step, 9) the source material is frozen at a temperature of −60° C. or below prior to or during the comminuting step, 10) the source material is frozen at a temperature of −80° C. or below prior to or during the comminuting step, 11) the mixing step is performed by blending or stirring, 12) the mixing step is performed by stirring, 13) the soluble collagen fraction is not filtered between the step of extracting the collagen from the insoluble fraction to form the soluble collagen fraction and the step of precipitating the collagen from the soluble collagen fraction, 14) the method further comprises the step of lyophilizing the collagen precipitate, 15) the method further comprises the step of polymerizing the collagen prior to or after lyophilization, 16) the collagen is precipitated by dialysis against a buffered solution, and 17) the dialysis tubing has a molecular weight cut-off of about 12,000 to about 14,000. In an alternative embodiment, any of these steps can be used in any combination.
  • In one embodiment, a method for engineering matrices with enhanced polymerization characteristics is provided. The method comprises the steps of obtaining collagen oligomers, polymerizing the collagen oligomers, and forming the engineered matrices with enhanced polymerization characteristics. An engineered matrix prepared by this method is also provided.
  • In other embodiments, the enhanced polymerization characteristics are selected from the group consisting of an enhanced rate of polymerization, a reduced lag time for polymerization, and an enhanced mechanical integrity, and the enhanced mechanical integrity is selected from the group consisting of strength and stiffness. In yet another embodiment, the collagen oligomers polymerize with a maximal t ½ selected from the group consisting of about 3 minutes, about 2.5 minutes, about 2.0 minutes, about 1.5 minutes, and about 1 minute.
  • In other illustrative embodiments, the collagen oligomers are obtained by isolation from a collagen-containing source material that is a tissue naturally enriched with collagen oligomers, the collagen oligomers are obtained by isolating and then chemically cross-linking collagen, or the collagen oligomers are isolated from a diseased tissue or a genetically-modified tissue.
  • In still another embodiment, an engineered matrix is provided. The matrix comprises a predetermined percentage of collagen oligomers based on total isolated collagen used to make the engineered matrix. In various embodiments, the collagen is isolated from a tissue naturally enriched with collagen oligomers, the collagen oligomers are isolated from a diseased tissue or a genetically-modified tissue, or the collagen oligomers are obtained by isolating and then chemically cross-linking collagen. In another illustrative embodiment, the predetermined percentage of collagen oligomers is selected from the group consisting of about 0.5% to about 100%, about 1.0% to about 100%, about 2% to about 100%, about 3% to about 100%, about 5% to about 100%, about 10% to about 100%, about 15% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 90% to about 100%, about 95% to about 100%, and about 100%. In still another embodiment, the engineered matrix further comprises a predetermined percentage of isolated collagen monomers.
  • In another aspect, a graft composition is provided. The graft composition comprises an engineered matrix comprising collagen oligomers wherein the matrix has a predetermined percentage of collagen oligomers based on total isolated collagen used to make the engineered matrix. In one embodiment, the predetermined percentage of collagen oligomers is selected from the group consisting of about 0.5% to about 100%, about 1.0% to about 100%, about 2% to about 100%, about 3% to about 100%, about 5% to about 100%, about 10% to about 100%, about 15% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 90% to about 100%, about 95% to about 100%, and about 100%. In various embodiments, the collagen oligomers are isolated from a diseased tissue or a genetically-modified tissue, the collagen oligomers are obtained by isolating and then chemically cross-linking collagen, or the collagen oligomers are isolated from a natural tissue enriched with collagen oligomers. In yet another embodiment, the graft composition further comprises a predetermined percentage of collagen monomers based on total isolated collagen used to make the engineered matrix.
  • Surprisingly, the inclusion of increased amounts of collagen oligomers in a collagen composition may increase the rate of polymerization, facilitate hierarchical assembly of component collagen fibrils, and enhance mechanical properties (e.g., strength and stiffness).
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows a polyacrylamide gel electrophoresis comparison of collagen isolated from pig skin, using the methods described herein, and a commercially available collagen preparation.
  • FIG. 2 shows a comparison by using turbidity analysis of the kinetics of polymerization of collagen isolated from pig skin using the methods described herein and a commercially available collagen preparation.
  • FIG. 3 shows a comparison using confocal reflection microscopy of the microstructure of an engineered ECM prepared from pig skin collagen, using the methods described herein, and a commercially available collagen preparation.
  • FIG. 4 shows a comparison using scanning electron microscopy of the microstructure of an engineered ECM prepared from pig skin collagen, using the methods described herein, and a commercially available collagen preparation.
  • FIG. 5 shows a comparison using a compression test of the mechanical properties (e.g., strength and modulus (stiffness)) of an engineered ECM prepared from pig skin collagen, using the methods described herein, and a commercially available collagen preparation.
  • FIG. 6 shows a polyacrylamide gel electrophoresis comparison of cyanogen bromide peptides from collagen isolated from pig skin collagen, using the methods described herein, and commercially available collagen preparations.
  • FIG. 7 shows the shear storage (G′) modulus during polymerization of an engineered ECM prepared from pig skin collagen, using the methods described herein, compared to a commercially available collagen preparation.
  • FIGS. 8A and B show the growth of mesenchymal stem cells on an engineered collagen ECM polymerized at 0.5 mg/ml (FIG. 8A) or 3 mg/ml (FIG. 8B) of collagen, and polymerized from pig skin collagen, isolated according to the methods described herein.
  • FIG. 9 shows the time-dependent glycerol-based separation of pig skin collagen type I oligomers and monomers. At each time point, the supernatant and pellet represent the monomer-rich and oligomer-rich fractions, respectively.
  • FIGS. 10A and B show glycerol-based separation of pig skin collagen type I oligomers and monomers. Pig skin collagen was polymerized in the presence of glycerol for 7 days. The mixture was centrifuged and the supernatant and pellet retained, dialyzed against dilute acetic acid and lyophilized to dryness. The supernatant and pellet represented monomer- and oligomer-rich collagen compositions, respectively, as determined by SDS-PAGE (4%). FIG. 10A shows the SDS-PAGE gel and FIG. 10B shows the corresponding densitometry analysis.
  • FIG. 11 shows SDS-PAGE (12%) analysis of CNBr peptide maps of collagen preps in which oligomer:monomer content is varied. Upon CNBr digestion, collagen preps yield distinct peptide fingerprints indicating distinct molecular compositions and cross-linked moieties.
  • FIGS. 12 A-B show the oligomer:monomer content of pig skin collagen preparation affects polymerization (self-assembly) kinetics. An increase in oligomer content decreases lag time (FIG. 12 B) and increases the rate of polymerization (FIG. 12 A). FIG. 12A shows A405 vs. time.
  • FIG. 13 shows the comparison of mechanical behavior of 3D engineered ECMs prepared with pig skin collagen preps of varied monomer:oligomer content. The shear storage modulus (G′) provides a measure of the stiffness of solid (elastic) components. Results show that G′ or stiffness of matrices increases with oligomer content.
  • FIG. 14 shows the comparison of mechanical behavior of 3D engineered ECMs prepared with pig skin collagen preps of varied monomer:oligomer content. An increase in delta as a function of strain as observed with the monomer-rich preparation indicates more fluid-like (viscous) behavior.
  • FIG. 15 shows the comparison of mechanical behavior of 3D engineered ECMs prepared with pig skin collagen preps of varied monomer:oligomer content. Matrices produced with collagen preps consisting of higher oligomer content show increased compressive modulus (stiffness) and failure strength.
  • FIG. 16 shows the compressive modulus (stiffness) values as a function of the oligomer:monomer content of the pig skin collagen preparation.
  • FIG. 17 shows the effects of oligomer content on the 3D collagen fibril microstructure of engineered ECMs.
  • FIG. 18 shows the effects of oligomer content on the 3D collagen fibril microstructure of engineered ECMs.
  • FIG. 19 shows the compositional analysis of different collagen preparations as determined using SDS-PAGE (4%). Results show that the pig skin collagen preparation uniquely contains collagen oligomers that run intermediate between commonly identified dimeric (β) and trimeric (γ) forms of collagen. Upon pepsin treatment of pig skin collagen, the content of unique collagen oligomers as other higher molecular weight collagen species is significantly reduced. Lane 1=PS+pepsin (acid solubilized type I collagen from pig skin posted-treated with pepsin); Lane 2=Vitrogen (pepsin solubilized type I collagen from calf skin); Lane 3=PureCol (pepsin solubilized type I collagen from calf skin); Lane 4=(Sigma, A) acid solubilized type I collagen from calf skin, Lot A; Lane 5=(Sigma, B) acid solubilized type I collagen from calf skin, Lot B; Lane 6=PS (acid solubilized type I collagen from pig skin).
  • FIG. 20 shows the kinetics of polymerization for the different collagen preparations as determined using spectrophotometric-based turbidity assay. Results show that the polymerization kinetics of collagen is highly dependent upon the collagen preparation. More specifically, polymerization of pig skin collagen shows a decreased lag phase and an increased rate of polymerization (slope of growth phase).
  • FIG. 21 shows the comparison of final absorbance (405 nm) values for different collagen preparations as determined from spectrophometric based turbidity assay. Results represent average of last 10 absorbance values.
  • FIG. 22 shows the collagen fibril microstructure of 3D engineered ECMs prepared with different purified type I collagen sources. Each collagen preparation was polymerized at two different concentrations (0.5 mg/ml and 2 mg/ml) using the same reaction conditions. Images represent 2D projections of 3D image stacks as collected using confocal reflection microscopy. Results show that the collagen fibril microstructure of 3D engineered ECMs is highly dependent upon the collagen preparation.
  • FIG. 23 shows the comparison of mechanical behavior 3D engineered ECMs prepared with different collagen preparations and tested in oscillatory shear. The storage modulus (G′) provides a measure of the stiffness of the solid (elastic) components. Results show that G′ or stiffness of matrices prepared with pig skin collagen are significantly greater than those obtained with the commercial collagen preparations (PureCol and Sigma).
  • FIGS. 24A-B show a comparison of collagen sources. FIG. 24A shows microstructural analysis of engineered ECMs using confocal reflection microscopy (fibril density vs. collagen concentration). FIG. 24B shows microstructural analysis of engineered ECMs using confocal reflection microscopy (average fibril diameter vs. collagen concentration).
  • DETAILED DESCRIPTION OF THE ILLUSTRATIVE EMBODIMENTS
  • As used herein, the term “lyophilized” means that water is removed from the composition, typically by freeze-drying under a vacuum. However, lyophilization can be performed by any method known to the skilled artisan and the method is not limited to freeze-drying under a vacuum.
  • As used herein “collagen-based matrix” means a matrix that comprises collagen. In illustrative embodiments, the “collagen-based matrices” described herein can be engineered from isolated collagen.
  • As used herein “engineered matrix” means a collagen-based matrix that is polymerized under conditions that are systematically varied where the conditions are selected from the group consisting of, but not limited to, pH, phosphate concentration, temperature, buffer composition, ionic strength, and composition and concentration of collagen.
  • As used herein, “isolated collagen” means any type of collagen, naturally present in a collagen-containing source material (described below) wherein the collagen has been at least partially purified by isolation and removal from the collagen-containing source material.
  • As used herein “sterilization” or “sterilize” or “sterilized” means removing unwanted contaminants including, but not limited to, endotoxins, nucleic acid contaminants, and infectious agents.
  • As used herein collagen “oligomer(s)” means covalently cross-linked collagen monomers (e.g., dimers=2 monomers, trimers=3 monomers, etc.).
  • The present invention relates to a method of preparing a collagen-based matrix. In one illustrative embodiment, methods for isolating collagen and then preparing the collagen-based matrix are provided. The method for isolating collagen comprises the steps of obtaining a collagen-containing source material, comminuting the source material, mixing the comminuted source material with an extraction solution, extracting the comminuted source material to form a soluble fraction and an insoluble fraction, obtaining the insoluble fraction, extracting the collagen from the insoluble fraction to form a soluble collagen fraction, precipitating the collagen from the soluble collagen fraction, and resuspending the precipitate in an aqueous solution wherein the aqueous solution used to resuspend the collagen precipitate is an acidic solution. In additional embodiments, the isolated collagen can be precipitated by dialysis against a buffered solution, for example, using dialysis tubing with a molecular weight cut-off of about 12,000 to about 14,000. In yet another embodiment, the isolated collagen can be lyophilized after precipitation.
  • In another illustrative aspect, a polymerizing step can be performed under conditions that are systematically varied where the conditions are selected from the group consisting of pH, phosphate concentration, temperature, buffer composition, ionic strength, the specific isolated collagen components present, and the concentration of the isolated collagen components present. In one illustrative embodiment, the isolated collagen can be lyophilized prior to polymerization. The isolated collagen can be lyophilized in an acid, such as acetic acid, hydrochloric acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid. In still another embodiment, the isolated collagen can be polymerized after precipitation, and optionally, the engineered matrix that results can be lyophilized.
  • In another illustrative embodiment, the invention relates to a collagen-based matrix prepared using the methods described above. In various embodiments, the collagen-based matrix can contain fibrils with specific characteristics, including, but not limited to, a fibril area fraction or a fibril volume fraction (i.e., density) of about 0.1% to about 100%, about 0.5% to about 100%, about 0.5% to about 26%, about 1% to about 100%, about 1% to about 26%, about 1% to about 7%, about 1% to about 15%, about 7% to about 26%, about 20% to about 30%, about 20% to about 50%, about 20% to about 70%, and about 20% to about 100%, about 30% to about 50%, about 30% to about 70%, and about 30% to about 100% and/or a modulus (e.g., an elastic or linear modulus, a compressive modulus, or a shear storage modulus) of about 0.5 kPa to about 40 kPa, about 30 kPa to 100 kPa, about 30 kPa to about 1000 kPa, about 30 kPa to about 10000 kPa, about 30 kPa to about 70000 kPa, about 100 kPa to 1000 kPa, about 100 kPa to about 10000 kPa, and about 100 kPa to about 70000 kPa.
  • Exemplary of tissues useful as a collagen-containing source material for isolating collagen to make the collagen-based matrices described herein are submucosa tissues or any other extracellular matrix-containing tissues of a warm-blooded vertebrate. Exemplary methods of preparing submucosa tissues are described in U.S. Pat. Nos. 4,902,508; 5,281,422; and 5,275,826, each incorporated herein by reference. Extracellular matrix material-containing tissues other than submucosa tissue may be used in accordance with the methods and compositions described herein. Methods of preparing other extracellular matrix material-derived tissues are known to those skilled in the art. For example, see U.S. Pat. Nos. 5,163,955 (pericardial tissue); 5,554,389 (urinary bladder submucosa tissue); 6,099,567 (stomach submucosa tissue); 6,576,265 (extracellular matrix tissues generally); 6,793,939 (liver basement membrane tissues); and U.S. patent application publication no. US-2005-0019419-A1 (liver basement membrane tissues); and international publication no. WO 2001/45765 (extracellular matrix tissues generally), each incorporated herein by reference. In various other embodiments, the collagen-containing source material can be selected from the group consisting of placental tissue, ovarian tissue, animal tail tissue, and skin tissue (e.g., see Example 1 and Gallop, et al., Preparation and Properties of Soluble Collagens, Meth. Enzymol. 6: 635-641 (1963), incorporated herein by reference). Any suitable extracellular matrix-containing tissue can be used as a collagen-containing source material.
  • In one illustrative embodiment, the extracellular matrix material-derived tissues used as the collagen-containing source material can be derived from a vertebrate tissue material comprising submucosa. In various embodiments, vertebrate tissue material comprising submucosa can be obtained from intestinal tissue harvested from animals raised for meat production, including, for example, pigs, cattle and sheep or other warm-blooded vertebrates. In various aspects, the tissue material comprising submucosa can be obtained from intestine, stomach, urinary bladder, the uterus, and any other submucosa-containing tissue.
  • An illustrative preparation method for tissue material comprising submucosa is described in U.S. Pat. No. 4,902,508, the disclosure of which is incorporated herein by reference. In one embodiment, a segment of vertebrate intestine, for example, preferably harvested from porcine, ovine or bovine species, but not excluding other species, is subjected to abrasion using a longitudinal wiping motion to remove cells. In this embodiment, the tissue material comprising submucosa is rinsed under hypotonic conditions, such as with water or with saline under hypotonic conditions and is optionally sterilized. In another illustrative embodiment, such compositions can be prepared by mechanically removing the luminal portion of the tunica mucosa and the external muscle layers and/or lysing resident cells with hypotonic washes, such as water or saline under hypotonic conditions. In these embodiments, the tissue material comprising submucosa can be stored in a hydrated or dehydrated state prior to extraction. In various aspects, the tissue material comprising submucosa can comprise any delamination embodiment, including the tunica submucosa delaminated from both the tunica muscularis and at least the luminal portion of the tunica mucosa of a warm-blooded vertebrate.
  • In various embodiments, the isolated collagen can also contain glycoproteins, proteoglycans, glycosaminoglycans (e.g., chondroitins and heparins), etc. extracted from the insoluble fraction with the collagen. The engineered matrices prepared by the methods described herein can serve as matrices for the regrowth of endogenous tissues at the implantation site (e.g., biological remodeling) which can assume the characterizing features of the tissue(s) with which they are associated at the site of implantation, insertion, or injection.
  • In various illustrative embodiments, the collagen-containing source material, the isolated collagen, or the collagen-based matrix, including an engineered matrix, can be disinfected and/or sterilized using conventional sterilization techniques including glutaraldehyde tanning, formaldehyde tanning at acidic pH, propylene oxide or ethylene oxide treatment, gas plasma sterilization, gamma radiation, electron beam, and/or peracetic acid sterilization. Sterilization techniques which do not adversely affect the structure and biotropic properties of the source material or the collagen can be used. Illustrative sterilization techniques are exposing the collagen-containing source material, the isolated collagen, or the collagen-based matrix, including an engineered matrix, to peracetic acid, 1-4 Mrads gamma irradiation (or 1-2.5 Mrads of gamma irradiation), ethylene oxide treatment, or gas plasma sterilization. In one embodiment, the collagen-containing source material, the isolated collagen, or the collagen-based matrix, including an engineered matrix, can be subjected to one or more sterilization processes. In an illustrative embodiment, peracetic acid can be used for sterilization.
  • Typically, prior to extraction, the collagen-containing source material is comminuted by tearing, cutting, grinding, or shearing the collagen-containing source material. In one illustrative embodiment, the collagen-containing source material can be comminuted by shearing in a high-speed blender, or by grinding the collagen-containing source material in a frozen state (e.g., at a temperature of −20° C., −40° C., −60° C., or −80° C. or below prior to or during the comminuting step) and then lyophilizing the material to produce a powder having particles ranging in size from about 0.1 mm2 to about 1.0 mm2. In one illustrative embodiment, the collagen-containing source material is comminuted by freezing and pulverizing under liquid nitrogen in an industrial blender. In this embodiment, the collagen-containing source material can be frozen in liquid nitrogen prior to, during, or prior to and during the comminuting step.
  • In one illustrative embodiment, after comminuting the collagen-containing source material, the material is mixed (e.g., by blending or stirring) with an extraction solution to extract and remove soluble proteins. Illustrative extraction solutions include sodium acetate (e.g., 0.5 M and 1.0 M). Other exemplary methods for extracting soluble proteins are known to those skilled in the art and are described in detail in U.S. Pat. No. 6,375,989, incorporated herein by reference. Illustrative extraction excipients include, for example, chaotropic agents such as urea, guanidine, sodium chloride or other neutral salt solutions, magnesium chloride, and non-ionic or ionic surfactants.
  • In one illustrative aspect, after the initial extraction, the soluble fraction can be separated from the insoluble fraction to obtain the insoluble fraction. For example, the insoluble fraction can be separated from the soluble fraction by centrifugation (e.g., 2000 rpm at 4° C. for 1 hour). In alternative embodiments, other separation techniques known to those skilled in the art, such as filtration, can be used. In one embodiment, the initial extraction step can be repeated one or more times, discarding the soluble fractions. In another embodiment, after completing the extractions, one or more steps can be performed of washing with water the insoluble fraction, followed by centrifugation, and discarding of the supernatant where the water is the supernatant.
  • In accordance with one illustrative embodiment, the insoluble fraction can then be extracted (e.g., with 0.075 M sodium citrate) to obtain the isolated collagen. In illustrative aspects the extraction step can be repeated multiple times retaining the soluble fractions. In one embodiment, the accumulated soluble fractions can be combined and can be clarified to form the soluble fraction, for example by centrifugation (e.g., 2000 rpm at 4° C. for 1 hour).
  • In one embodiment, the soluble fraction can be fractionated to precipitate the isolated collagen. In one illustrative aspect, the soluble fraction can be fractionated by dialysis. Exemplary molecular weight cut-offs for the dialysis tubing or membrane are from about 3,500 to about 12,000 or about 3,500 to about 5,000 or about 12,000 to about 14,000. In various illustrative embodiments, the fractionation, for example by dialysis, can be performed at about 2° C. to about 37° C. for about 1 hour to about 96 hours. In one embodiment, the soluble fraction is dialyzed against a buffered solution (e.g., 0.02 M sodium phosphate dibasic). However, the fractionation can be performed at any temperature, for any length of time, and against any suitable buffered solution. In one embodiment, the precipitated collagen is then collected by centrifugation (e.g., 2000 rpm at 4° C. for 1 hour). In another embodiment, after precipitation, one or more steps can be performed of washing the precipitate with water, followed by centrifugation, and discarding of the supernatant where the water is the supernatant.
  • In various illustrative embodiments, the precipitated collagen can then be resuspended in an aqueous solution wherein the aqueous solution is acidic. For example, the aqueous acidic solution can be an acetic acid solution, but any other acids including hydrochloric acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid can be used. For example, acids, at concentrations of from about 0.001 N to about 0.1 N, from about 0.005 N to about 0.1 N, from about 0.01 N to about 0.1 N, from about 0.05 N to about 0.1 N, from about 0.001 N to about 0.05 N, from about 0.001 N to about 0.01 N, or from about 0.01 N to about 0.05 N can be used to resuspend the precipitate.
  • As discussed above, the term “lyophilized” means that water is removed from the composition, typically by freeze-drying under a vacuum. In one illustrative aspect, the isolated resuspended collagen can be lyophilized after it is resuspended. In another illustrative embodiment, the polymerized matrix itself can be lyophilized. In one illustrative lyophilization embodiment, the resuspended collagen is first frozen, and then placed under a vacuum. In another lyophilization embodiment, the resuspended collagen can be freeze-dried under a vacuum. In another lyophilization embodiment, the precipitated collagen can be lyophilized before resuspension. Any method of lyophilization known to the skilled artisan can be used.
  • In additional embodiments, the acids described above can be used as adjuvants for storage after lyophilization in any combination. The acids that can be used as adjuvants for storage include hydrochloric acid, acetic acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid, and these acids can be used at any of the above-described concentrations. In one illustrative embodiment, the lyophilizate can be stored (e.g., lyophilized in and stored in) an acid, such as acetic acid, at a concentration of from about 0.001 N to about 0.5 N or from about 0.01 N to about 0.5 N. In another embodiment, the lyophilizate can be stored in water with a pH of about 6 or below. In another embodiment, the lyophilized product can be stored dry. In other illustrative embodiments, lyoprotectants, cryoprotectants, lyophilization accelerators, or crystallizing excipients (e.g., ethanol, isopropanol, mannitol, trehalose, maltose, sucrose, tert-butanol, and tween 20), or combinations thereof, and the like can be present during lyophilization.
  • In accordance with one illustrative embodiment, the resuspended collagen is sterilized. Exemplary sterilizing and/or disinfecting agents are described above, but any sterilizing and/or disinfecting agent or method of sterilization known in the art can be used. The resuspended collagen can be sterilized using chloroform, glutaraldehyde, formaldehyde, acidic pH, propylene oxide, ethylene oxide, gas plasma sterilization, gamma radiation, electron beam sterilization, or peracetic acid sterilization, or combinations thereof, and the like. Illustrative sterilization techniques are exposing the resuspended collagen to peracetic acid, 1-4 Mrads gamma irradiation (or 1-2.5 Mrads of gamma irradiation), ethylene oxide treatment, or gas plasma sterilization.
  • In one embodiment, the isolated collagen can be sterilized before lyophilization. In another illustrative embodiment the isolated collagen can be sterilized after lyophilization or the collagen-containing source material can be sterilized. Sterilization of the collagen-containing source material can be performed, for example, as described in U.S. Pat. Nos. 4,902,508 and 6,206,931, incorporated herein by reference. In another illustrative embodiment, the polymerized matrix formed from the isolated collagen is sterilized.
  • In one illustrative embodiment, the isolated collagen is directly sterilized after resuspension, for example, with peracetic acid or with peracetic acid and ethanol (e.g., by the addition of 0.18% peracetic acid and 4.8% ethanol to the resuspended collagen solution before lyophilization). In another embodiment, sterilization can be carried out during the fractionation step. For example, the isolated collagen composition can be dialyzed against chloroform, peracetic acid, or a solution of peracetic acid and ethanol to disinfect or sterilize the isolated collagen. Illustratively, the isolated collagen can be sterilized by dialysis against a solution of peracetic acid and ethanol (e.g., 0.18% peracetic acid and 4.8% ethanol). The chloroform, peracetic acid, or peracetic acid/ethanol can be removed prior to lyophilization, for example by dialysis against an acid, such as 0.01 N acetic acid. In an alternative embodiment, the lyophilized composition can be sterilized directly after rehydration, for example, by the addition of 0.18% peracetic acid and 4.8% ethanol. In this embodiment, the sterilizing agent can be removed prior to polymerization of the isolated collagen to form fibrils.
  • If the isolated collagen or polymerized collagen is lyophilized, the lyophilized composition can be stored frozen, refrigerated, or at room temperature (for example, at about −80° C. to about 25° C.). Storage temperatures are selected to stabilize the collagen. The compositions can be stored for about 1-26 weeks, or longer.
  • In one embodiment, the isolated collagen can be dialyzed against 0.01 N acetic acid, for example, prior to lyophilization to remove the sterilization solution and so that the isolated collagen is in a 0.01 N acetic acid solution. In another embodiment, the isolated collagen can be dialyzed against hydrochloric acid, for example, prior to lyophilization and can be lyophilized in hydrochloric acid and redissolved in hydrochloric acid, acetic acid, or water.
  • If the isolated collagen is lyophilized, the resulting lyophilizate can be redissolved in any solution, but may be redissolved in an acidic solution or water. In various aspects, the lyophilizate can be redissolved in, for example, acetic acid, hydrochloric acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid, at any of the above-described concentrations, or can be redissolved in water. In one illustrative embodiment the lyophilizate is redissolved in 0.01 N acetic acid. For use in producing engineered matrices that can be injected in vivo or used for other purposes in vitro, the redissolved lyophilizate can be subjected to varying conditions (e.g., pH, phosphate concentration, temperature, buffer composition, ionic strength, and composition and concentration of the isolated collagen components (dry weight/ml)) that result in polymerization to form engineered matrices for specific tissue graft applications.
  • For use in producing engineered matrices that can be 1.) injected in vivo and used for specific tissue graft applications or 2.) used for other purposes in vitro, such as studying cell-extracellular matrix interactions, the isolated collagen can be subjected to varying conditions for polymerization to form fibrils. In illustrative embodiments, the conditions that can be varied include pH, phosphate concentration, temperature, buffer composition, ionic strength, the particular isolated collagen components (e.g., type I and type III collagen) and other ECM components present, and the concentration of the isolated collagen (dry weight/ml). These conditions result in polymerization of the isolated collagen to form engineered matrices with desired compositional, microstructural, and mechanical characteristics. Illustratively, these compositional, microstructural, and mechanical characteristics can include fibril length, fibril diameter, number of fibril-fibril connections (e.g., cross-links), fibril density, fibril organization, matrix composition, 3-dimensional shape or form, and viscoelastic, tensile, shear, or compressive behavior (e.g., failure stress, failure strain, and modulus), permeability, degradation rate, swelling, hydration properties (e.g., rate and swelling), and in vivo tissue remodeling and bulking properties, rate of polymerization, lag time of polymerization, extent of polymerization, viscosity of interstitial fluid and desired in vitro and in vivo cell responses. The collagen-based matrices described herein have desirable biocompatibility and in vitro and in vivo remodeling properties, among other desirable properties.
  • In various illustrative embodiments, qualitative and quantitative microstructural characteristics of the engineered matrices can be determined by environmental or cryostage scanning electron microscopy, transmission electron microscopy, confocal microscopy, second harmonic generation multi-photon microscopy. In another embodiment, polymerization kinetics may be determined by spectrophotometry or time-lapse confocal reflection microscopy. In another embodiment, tensile, compressive and viscoelastic properties can be determined by rheometry or tensile testing. In another embodiment, a rat subcutaneous injection model can be used to determine remodeling properties. All of these methods are known in the art or are further described in the Examples or are described in Roeder et al., J. Biomech. Eng., vol. 124, pp. 214-222 (2002), in Pizzo et al., J. Appl. Physiol., vol. 98, pp. 1-13 (2004), Fulzele et al., Eur. J. Pharm. Sci., vol. 20, pp. 53-61 (2003), Griffey et al., J. Biomed. Mater. Res., vol. 58, pp. 10-15 (2001), Hunt et al., Am. J. Surg., vol. 114, pp. 302-307 (1967), and Schilling et al., Surgery, vol. 46, pp. 702-710 (1959), incorporated herein by reference.
  • In accordance with one embodiment, the isolated collagen is polymerized to form fibrils at a final concentration (dry weight/ml) of about 0.05 to about 5.0 mg/ml of the isolated collagen, or in another embodiment the final concentration is selected from the range of about 0.05 mg/ml to about 4.0 mg/ml, and in another embodiment the final concentration is selected from the range of about 0.05 mg/ml to about 3.0 mg/ml, and in another embodiment the final concentration is about 0.05, 0.1, 0.2, 0.3, 0.5, 1.0, 2.0, or 3.0 mg/ml. In other embodiments, the isolated collagen is polymerized at final concentrations (dry weight/ml) of about 5 to about 10 mg/ml, about 5 to about 30 mg/ml, about 5 to about 50 mg/ml, about 5 to about 100 mg/ml, about 20 to about 50 mg/ml, about 20 to about 60 mg/ml, or about 20 to about 100 mg/ml.
  • In various illustrative embodiments, the polymerization reaction is conducted in a buffered solution using any biologically compatible buffer known to those skilled in the art. For example, the buffer may be selected from the group consisting of phosphate buffer saline (PBS), Tris(hydroxymethyl)aminomethane Hydrochloride (Tris-HCl), 3-(N-Morpholino) Propanesulfonic Acid (MOPS), piperazine-n,n′-bis(2-ethanesulfonic acid) (PIPES), [n-(2-Acetamido)]-2-Aminoethanesulfonic Acid (ACES), N-[2-hydroxyethyl]piperazine-N′-[2-ethanesulfonic acid] (HEPES), and 1,3-bis[tris(Hydroxymethyl)methylamino]propane (Bis Tris Propane). In one embodiment the buffer is PBS, Tris, or MOPS and in one embodiment the buffer system is PBS.
  • In various illustrative embodiments, the polymerization of the isolated collagen is conducted at a pH selected from the range of about 5.0 to about 11, and in one embodiment polymerization is conducted at a pH selected from the range of about 6.0 to about 9.0, and in one embodiment polymerization is conducted at a pH selected from the range of about 6.5 to about 8.5, and in another embodiment the polymerization of the isolated collagen is conducted at a pH selected from the range of about 7.0 to about 8.5, and in another embodiment the polymerization of the isolated collagen is conducted at a pH selected from the range of about 7.3 to about 7.4.
  • In other illustrative aspects, the ionic strength of the buffered solution is also regulated. In accordance with one embodiment, the ionic strength of the buffer used to polymerize the isolated collagen is selected from a range of about 0.05 to about 1.5 M, in another embodiment the ionic strength is selected from a range of about 0.10 to about 0.90 M, in another embodiment the ionic strength is selected from a range of about 0.14 to about 0.30 M and in another embodiment the ionic strength is selected from a range of about 0.14 to about 0.17 M.
  • In still other illustrative embodiments, the polymerization is conducted at temperatures selected from the range of about 0° C. to about 60° C. In other embodiments, the polymerization is conducted at temperatures above 20° C., and typically the polymerization is conducted at a temperature selected from the range of about 20° C. to about 40° C., and more typically the temperature is selected from the range of about 30° C. to about 40° C. In one illustrative embodiment the polymerization is conducted at about 37° C.
  • In yet other embodiments, the phosphate concentration is varied. For example, in one embodiment, the phosphate concentration is selected from a range of about 0.005 M to about 0.5 M. In another illustrative embodiment, the phosphate concentration is selected from a range of about 0.01 M to about 0.2 M. In another embodiment, the phosphate concentration is selected from a range of about 0.01 M to about 0.1 M. In another illustrative embodiment, the phosphate concentration is selected from a range of about 0.01 M to about 0.03 M.
  • In other illustrative embodiments, the isolated collagen can be polymerized by, for example, extrusion into a desired buffer, including the buffers described above, or wet-spinning to form strands of isolated collagen. In one embodiment the strands can be formed by extrusion through a needle and can be air-dried to form fibers or threads of various dimensions. The syringe can be adapted with needles or tubing to control the dimensions (e.g., diameter) of the fibers or threads. In one embodiment, the extrusion process involves polymerization of the isolated collagen followed by extrusion into a bath containing water, a buffer, or an organic solvent (e.g., ethanol). In another embodiment, the extrusion process involves coextrusion of the isolated collagen with a polymerization buffer (e.g., the buffer such as Tris or phosphate buffers at various concentrations can be varied to control pH and ionic strength). In yet another embodiment, the extrusion process involves extrusion of the isolated collagen into a polymerization bath (e.g., the buffer such as Tris or phosphate buffers at various concentrations can be varied to control pH and ionic strength). The bath conditions affect polymerization time and properties of the fibers or threads, such as mechanical integrity of the fibers or threads, fiber dimensions, and the like. In one embodiment, the fibers can be air-dried to create materials suitable for use as sutures. Multiple fibers can be assembled (e.g., woven, braided to form matrix constructs).
  • In various embodiments, the engineered matrices of the present invention can be combined, prior to, during, or after polymerization, with nutrients, including minerals, amino acids, sugars, peptides, proteins, vitamins (such as ascorbic acid), or glycoproteins that facilitate cellular proliferation, such as laminin and fibronectin, hyaluronic acid, or growth factors such as epidermal growth factor, platelet-derived growth factor, transforming growth factor beta, or fibroblast growth factor, and glucocorticoids such as dexamethasone. In other illustrative embodiments, fibrillogenesis inhibitors, such as glycerol, glucose, or polyhydroxylated compounds can be added prior to or during polymerization. In accordance with one embodiment, cells can be added to the isolated collagen as the last step prior to the polymerization or after polymerization of the engineered matrix. In another illustrative embodiment, particulate extracellular matrix material can be added to the isolated collagen and can enhance bulking capacity. In other illustrative embodiments, cross-linking agents, such as carbodiimides, aldehydes, lysl-oxidase, N-hydroxysuccinimide esters, imidoesters, hydrazides, and maleimides, and the like can be added before, during, or after polymerization.
  • The isolated collagen is derived from a collagen-containing source material and, in some embodiments, may contain glycoproteins, such as laminin and fibronectin, proteoglycans, such as serglycin, versican, decorin, and perlecan, and glycosaminoglycans. In one embodiment, the isolated collagen can be further purified or partially purified and the purified or partially purified composition can be used in accordance with the methods described herein or mixtures of partially purified or purified components can be used. As used herein, the term “purified” means the isolation of collagen in a form that is substantially free from other components (e.g., typically the total amount of other components present in the composition represents less than 5%, or more typically less than 0.1%, of total dry weight).
  • In additional illustrative embodiments, engineered matrices are provided. The engineered matrices can be prepared according to the methods described herein, including any of the methods or conditions for polymerization described herein. The engineered matrices comprise collagen fibrils. As used herein the term “collagen fibril” refers to a quasi-crystalline, filamentous structure formed by the self-assembly of soluble collagen molecules. The engineered matrices comprise collagen fibrils which may pack in a quarter-staggered pattern giving the fibril a characteristic striated appearance or banding pattern along its axis. Collagen fibrils are distinct from the amorphous aggregates or precipitates of insoluble collagen that can be formed by dehydrating (e.g., lyophilizing) collagen suspensions to form porous network scaffolds.
  • Typically, the matrices are prepared from isolated collagen at collagen concentrations ranging from about 0.05 to about 5.0 mg/ml, about 1.0 mg/ml to about 3.0 mg/ml, about 0.05 mg/ml to about 10 mg/ml, about 0.05 to about 20 mg/ml, about 0.05 to about 30 mg/ml, about 0.05 to about 40 mg/ml, about 0.05 to about 50 mg/ml, about 0.05 to about 60 mg/ml, about 0.05 to about 80 mg/ml, about 5 mg/ml to 10 mg/ml, about 5 mg/ml to 20 mg/ml, about 5 mg/ml to about 40 mg/ml, about 5 mg/ml to 60 mg/ml, about 5 mg/ml to about 100 mg/ml, about 20 mg/ml to about 40 mg/ml, about 20 mg/ml to 60 mg/ml, or about 20 mg/ml to about 100 mg/ml.
  • In another illustrative embodiment, the engineered matrices contain fibrils with specific characteristics, including, but not limited to, a fibril area fraction (defined as the percent area of the total area occupied by fibrils in a cross-sectional surface of the matrix; 2-dimensional) or a fibril volume fraction (the percent area of the total area occupied by fibrils in 3 dimensions) of about 0.1% to about 100%, about 0.5% to about 100%, about 0.5% to about 26%, about 1% to about 100%, about 1% to about 26%, about 1% to about 7%, about 1% to about 15%, of about 7% to about 26%, about 20% to about 30%, about 20% to about 50%, about 20% to about 70%, about 20% to about 100%, about 30% to about 50%, about 30% to about 70%, or about 30% to about 100%.
  • In yet another embodiment, the three-dimensional engineered matrices have a modulus (e.g., an elastic or linear modulus (defined by the slope of the linear region of the stress-strain curve obtained using conventional mechanical testing protocols; i.e., stiffness), a compressive modulus, or a shear storage modulus) of about 0.5 kPa to about 40 kPa, about 30 kPa to 100 kPa, about 30 kPa to about 1000 kPa, about 30 kPa to about 10000 kPa, about 30 kPa to about 70000 kPa, about 100 kPa to 1000 kPa, about 100 kPa to about 10000 kPa, or about 100 kPa to about 70000 kPa.
  • In another embodiment the engineered matrices have a fibril area fraction or a fibril volume fraction of about 0.5% to about 1%, about 0.5% to about 5%, 0.5% to about 10%, about 0.5% to about 50%, about 0.5% to about 100%, or of about 7% to about 26%. In another embodiment the engineered matrices have a fibril area fraction or a fibril volume fraction of about 7% to about 15% or about 16% to about 26%. In another embodiment the engineered matrices have a fibril area fraction or a fibril volume fraction of about 18.5% to about 25%. In another embodiment the engineered matrices are formed from collagen at concentrations of about 3.2, 3.4, 3.6, 3.8, 4.0, 4.5 or 5.0 mg/ml of collagen, resulting in engineered matrices having a fibril area fraction of about 19%, 19.7%, 20.5%, 21.2%, 22%, 23.8% and 25.6%, respectively.
  • In yet another embodiment, the engineered matrices have a modulus of about 0.5 to about 40 kPa. In accordance with another embodiment, the engineered matrices have a relatively low modulus of about 0.5 to about 24.0 kPa. In one other embodiment, the engineered matrices have a relatively high modulus of about 25 to about 40 kPa.
  • In illustrative embodiments, as discussed above, the polymerization reaction for engineered matrices can be conducted in a buffered solution using any biologically compatible buffer system known to those skilled in the art. For example, the buffer may be selected from the group consisting of phosphate buffer saline (PBS), Tris (hydroxymethyl)aminomethane Hydrochloride (Tris-HCl), 3-(N-Morpholino) Propanesulfonic Acid (MOPS), piperazine-n,n′-bis(2-ethanesulfonic acid) (PIPES), [n-(2-Acetamido)]-2-Aminoethanesulfonic Acid (ACES), N-[2-hydroxyethyl]piperazine-N′-[2-ethanesulfonic acid] (HEPES) and 1,3-bis[tris(Hydroxymethyl)methylamino]propane (Bis Tris Propane). In one embodiment the buffer is PBS, Tris, or MOPS and in one embodiment the buffer system is PBS, and more particularly 10×PBS. In accordance with one embodiment, the 10×PBS buffer at pH 7.4 comprises the following ingredients:
  • 1.37 M NaCl
  • 0.027 M KCl
  • 0.081 M Na2HPO4
  • 0.015 M KH2PO4
  • 5 mM MgCl2
  • 55.5 mM glucose
  • In one embodiment, to create 10×PBS buffers of different pH's, the ratio of Na2HPO4 and KH2PO4 is varied. In another embodiment, ionic strength may be adjusted as an independent variable by varying the molarity of NaCl only.
  • In accordance with one embodiment, for polymerization to form the engineered matrices, the isolated collagen can be pipetted into a plate with wells and the isolated collagen can be allowed to polymerize under any of the conditions described above. For example, a humidified environment at 37° C. for approximately 30 minutes can be used. In an alternative embodiment, the isolated collagen is injected into a host and is polymerized in vivo.
  • As discussed above, in accordance with one embodiment, cells can be added to the isolated collagen as the last step prior to the polymerization. In another embodiment, cells can be added after polymerization of the engineered matrix. The engineered matrices comprising the cells can be subsequently injected or implanted in a host for use as a tissue graft. In another embodiment, the cells on or within the engineered matrix can be cultured in vitro, for a predetermined length of time, to increase the cell number or to induce desired remodeling prior to implantation or injection into a host. In a further embodiment, the cells can be cultured in vitro, for a predetermined length of time, to increase cell number and the cells can be separated from the matrix and implanted or injected into the host in the absence of the engineered matrix.
  • In still another illustrative embodiment, the engineered matrices can include exogenous glucose and/or calcium chloride in the interstitial fluid of the matrices to promote cell growth. In one embodiment, about 1.0 mM to about 300 mM glucose and about 0.2 mM to about 4.0 mM CaCl2 is included. In one embodiment, the isolated collagen comprises about 0.1 mg/ml to about 3 mg/ml total isolated collagen in about 0.05 to about 0.005N HCl, about 0.07M to about 0.28M NaCl, about 1.3 to about 4.5 mM KCl, about 4.0 to about 16 mM Na2HPO4, about 0.7 to about 3.0 mM KH2PO4, about 0.25 to about 1.0 mM MgCl2, and about 2.77 mM to about 166.5 mM glucose. In this embodiment, polymerization of the isolated collagen is induced by the addition of a neutralizing solution such as NaOH. For example, a NaOH solution can be added to a final concentration of 0.01N NaOH. In this embodiment, cells are then added and a calcium chloride solution is also added to bring the final concentration of CaCl2 to about 0.4 mM to about 2.0 mM CaCl2. The composition is then allowed to polymerize either in vitro or in vivo to form an engineered matrix comprising collagen fibrils with cells in and/or on the matrix.
  • In accordance with one embodiment, a kit is provided for preparing engineered matrices. In this embodiment, the kit comprises sterilized components that can be combined to form an engineered matrix comprising collagen fibrils. In one embodiment, cells may constitute a component of the kit. In accordance with one embodiment, the kit comprises an isolated collagen composition, and a polymerization composition. In one embodiment, the kit comprises separate vessels, each containing one of the following components: isolated collagen, a phosphate buffer solution, a glucose solution, a calcium chloride solution, and a basic neutralizing solution. In one embodiment, the isolated collagen is provided in a lyophilized form and the kit is further provided with a solution of acetic acid (or other dilute acid including for example, hydrochloric acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid) for resuspending the lyophilized isolated collagen.
  • In another embodiment, the kit comprises a solution comprising isolated collagen, a phosphate buffer solution, a glucose solution, a calcium chloride solution, an acid solution, and a basic neutralizing solution. In another embodiment, the kit can include isolated collagen and a polymerization buffer. In one embodiment the polymerization composition comprises a phosphate buffer that has a pH of about 7.2 to about 7.6 and the acid solution is an acetic acid solution comprising about 0.05N to about 0.005N acetic acid. In another embodiment, the acid solution is about 0.01N acetic acid. In another embodiment the polymerization composition comprises a phosphate buffer that has a pH of about 7.2 to about 7.6 and the acid solution is a hydrochloric acid solution comprising about 0.05N to about 0.005N hydrochloric acid. In another embodiment, the acid solution is about 0.01N hydrochloric acid. In one embodiment, the glucose solution has a concentration selected from the range of about 0.2% to about 5% w/v glucose, or about 0.5% to about 3% w/v glucose, and in one embodiment the glucose solution is about 1% w/v glucose. In one embodiment the CaCl2 solution has a concentration selected from the range of about 2 mM to about 40.0 mM CaCl2 or about 0.2 mM to about 4.0 mM CaCl2, or about 0.2 to about 2 mM CaCl2. In one embodiment the kit is provided with a 10×PBS buffer having a pH of about pH 7.4, and comprising about 1.37M NaCl, about 0.027M KCl, about 0.081M Na2HPO4, about 0.015M KH2PO4, about 5 mM MgCl2 and about 1% w/v glucose. In another embodiment, kits are provided that comprise three-dimensional, preformed engineered matrices prepared according to any of the methods described herein and wherein the kits comprise any of the components described herein.
  • The kits can further comprise instructional materials describing methods for mixing the kit reagents to prepare engineered matrices or describing methods for using preformed, three-dimensional engineered matrices. In particular, the instructional materials can provide information regarding the final concentrations that give optimal microenvironmental conditions including fibril microstructure and mechanical properties for a particular cell type or for a particular desired result.
  • The lyophilized isolated collagen prepared by the methods described herein maintains its bioactivity (i.e., the capacity to polymerize and form fibrils in vitro or in vivo and to remodel tissue in vivo). In one embodiment, the isolated collagen can be used, for example, for making engineered matrices for specific tissue graft applications. In another embodiment, the lyophilized isolated collagen is useful commercially for the mass production of isolated collagen (i.e., the components can be lyophilized and stored without loss of bioactivity) for use in making engineered matrices. Lyophilized, isolated collagen that retains bioactivity and polymerization capacity is also useful for the concentration of isolated collagen for preparing engineered matrices that require concentration (i.e., higher concentrations) of collagen.
  • Surprisingly, the inclusion of increased amounts of isolated collagen oligomers in a collagen composition may increase the rate of polymerization, decrease lag time, facilitate hierarchical assembly of component collagen fibrils, and enhance mechanical properties (e.g., strength and stiffness). Accordingly, in one embodiment, a collagen composition is provided comprising collagen oligomers wherein the oligomers are isolated from a mammalian tissue enriched with or containing collagen oligomers.
  • In another illustrative embodiment, a method for engineering matrices with enhanced polymerization characteristics is provided. The method comprises the steps of obtaining collagen oligomers, polymerizing the collagen oligomers, and forming the engineered matrices with enhanced polymerization characteristics. In another aspect, the enhanced polymerization characteristics are selected from the group consisting of an enhanced rate of polymerization, a reduced lag time for polymerization, and an enhanced mechanical integrity. In another embodiment, the enhanced mechanical integrity is selected from the group consisting of strength and stiffness. In another embodiment, the polymerization characteristics are enhanced relative to type I isolated collagen obtained from Sigma-Aldrich as described in Example 2 and polymerized under the conditions described in Example 3. In still another embodiment, the collagen oligomers polymerize with a maximal T ½ of 3 minutes, 2.5 minutes, 2.0 minutes, 1.5 minutes, or 1 minute.
  • In yet another embodiment, the collagen-containing source material enriched with collagen oligomers is a tissue naturally enriched with collagen oligomers. In other embodiments, the collagen-containing source material enriched with collagen oligomers is a diseased tissue or a genetically-modified tissue where the tissue is enriched with collagen oligomers. In another illustrative aspect, the collagen-containing source material enriched with collagen oligomers is a mechanically-modified tissue or an electrically-modified tissue. In another embodiment, the collagen-containing source material is mechanically modified cultured cells or electrically-modified cultured cells. In still another aspect, the collagen oligomers are obtained by isolating and then chemically cross-linking collagen.
  • Extracellular matrix materials can be utilized as tissue graft compositions for remodeling tissues in vivo or for in vitro applications. The methods of collagen isolation and the collagen compositions described herein allow for the controlled alteration of the microstructural and subsequent mechanical properties of a resulting engineered matrix for such uses as graft compositions for inducing the repair of damaged or diseased tissues in vivo, and for inducing the proliferation or other fundamental behaviors of cells in vitro. Systematic variation of engineered matrix design features using the methods of polymerization described herein provides control of in vivo (e.g., cell infiltration, vascularization, rate of cell proliferation, differentiation, morphogenesis, remodeling, degradation, and contractility) and in vitro (e.g., cell morphology, migration, proliferation, differentiation, morphogenesis, and contractility) responses.
  • Accordingly, in another embodiment a graft composition comprising an engineered matrix comprising collagen oligomers is provided. The graft composition has a predetermined percentage of collagen oligomers based on total isolated collagen added to make the engineered matrix. In various embodiments, the predetermined percentage of collagen oligomers can be about 0.5% to about 100%, about 1% to about 100%, about 2% about 100%, about 3% to about 100%, about 5% to about 100%, about 10% to about 100%, about 15% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 90% to about 100%, about 95% to about 100%, or about 100%. In yet another embodiment, the collagen oligomers are obtained from a collagen-containing source material enriched with collagen oligomers (e.g., pig skin). In other embodiments, the collagen-containing source material enriched with collagen oligomers is a natural tissue, a diseased tissue or a genetically-modified tissue where the tissue is enriched with collagen oligomers. In still another aspect, the graft composition comprises collagen oligomers obtained by isolating and then chemically cross-linking collagen.
  • In another embodiment, the polymerization characteristics of the engineered matrix used to make the graft composition are enhanced relative to type I isolated collagen obtained from Sigma-Aldrich as described in Example 2 and polymerized under the conditions described in Example 3. In still another embodiment, the collagen oligomers that polymerize to form the engineered matrix polymerize with a maximal T ½ of 3 minutes, 2.5 minutes, 2.0 minutes, 1.5 minutes, or 1 minute.
  • All of the disclosure herein of methods of isolation of collagen, methods of polymerization of isolated collagen to form engineered matrices, the use of those engineered matrices in graft compositions, and disclosure relating to isolated collagen compositions and graft compositions, applies equally to isolated collagen enriched with collagen oligomers. In one illustrative embodiment, isolated collagen enriched with oligomers can be prepared by isolating collagen from a natural tissue (e.g., pig skin) according to the methods described herein. In one embodiment, the collagen oligomers can be isolated by a method such as the method described in Example 10. In other embodiments, oligomer-monomer separation can be achieved using methods that vary polymerization reaction parameters including temperature, pH, and ionic strength. Any other method known to the skilled artisan to be useful for separating collagen oligomers and monomers can be used.
  • In accordance with one embodiment, a kit is provided for preparing engineered matrices. In this embodiment, the kit comprises sterilized components that can be combined to form an engineered matrix comprising collagen fibrils. In one embodiment, the sterilized components include isolated collagen oligomers. In one embodiment, cells may constitute a component of the kit. In accordance with one embodiment, the kit comprises an isolated collagen oligomer composition, and a polymerization composition. In one embodiment, the kit comprises separate vessels, each containing one of the following components: isolated collagen oligomers, a phosphate buffer solution, a glucose solution, a calcium chloride solution, and a basic neutralizing solution. In another embodiment, the kit comprises separate vessels, each containing one of the following components: isolated collagen oligomers, isolated collagen monomers, a phosphate buffer solution, a glucose solution, a calcium chloride solution, and a basic neutralizing solution. In one embodiment, the isolated collagen is provided in a lyophilized form and the kit is further provided with a solution of acetic acid (or other dilute acid including for example, hydrochloric acid, formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid) for resuspending the lyophilized isolated collagen oligomers or monomers.
  • In another embodiment, the kit comprises a solution comprising isolated collagen oligomers and/or monomers, a phosphate buffer solution, a glucose solution, a calcium chloride solution, an acid solution, and a basic neutralizing solution. In another embodiment, the kit can comprise isolated collagen and a polymerization buffer. In one embodiment the polymerization composition comprises a phosphate buffer that has a pH of about 7.2 to about 7.6 and the acid solution is an acetic acid solution comprising about 0.05N to about 0.005N acetic acid. In another embodiment, the acid solution is about 0.01N acetic acid. In one embodiment the polymerization composition comprises a phosphate buffer that has a pH of about 7.2 to about 7.6 and the acid solution is a hydrochloric acid solution comprising about 0.05N to about 0.005N hydrochloric acid. In another embodiment, the acid solution is about 0.01N hydrochloric acid. In one embodiment, the glucose solution has a concentration selected from the range of about 0.2% to about 5% w/v glucose, or about 0.5% to about 3% w/v glucose, and in one embodiment the glucose solution is about 1% w/v glucose. In one embodiment the CaCl2 solution has a concentration selected from the range of about 2 mM to about 40.0 mM CaCl2 or about 0.2 mM to about 4.0 mM CaCl2, or about 0.2 to about 2 mM CaCl2. In one embodiment the kit is provided with a 10×PBS buffer having a pH of about pH 7.4, and comprising about 1.37M NaCl, about 0.027M KCl, about 0.081M Na2HPO4, about 0.015M KH2PO4, about 5 mM MgCl2 and about 1% w/v glucose. In another embodiment, kits are provided that comprise three-dimensional, preformed engineered matrices prepared according to any of the methods described herein, including engineered matrices formed from isolated collagen oligomers alone, or isolated collagen oligomers and monomers in predetermined percentages of total collagen used to form the matrix, and wherein the kits comprise any of the components described herein.
  • The kits can further comprise instructional materials describing methods for mixing the kit reagents to prepare engineered matrices or describing methods for using preformed, three-dimensional engineered matrices. In particular, the instructional materials can provide information regarding the final concentrations that give optimal microenvironmental conditions including fibril microstructure and mechanical properties for a particular cell type or for a particular desired result.
  • The following examples illustrate specific embodiments in further detail. These examples are provided for illustrative purposes only and should not be construed as limiting the invention or the inventive concept in any way.
  • EXAMPLE 1 Collagen Isolation
  • To prepare collagen, skin was harvested from pig immediately following euthanasia and was washed thoroughly with cold water. The skin was stretched out and pinned to a board and stored at 4° C. The hair was removed with clippers. The dermal layer of the tissue was isolated by separating and removing the upper epidermal layer and the lower loose fatty connective layers. This removal was readily achieved by scraping the tissue with a knife or straight razor. The tissue was maintained at 4° C.
  • The resulting dermal layer tissue was washed in water and then cut into small pieces (approximately 1 cm2) and was frozen and stored at −80° C. The frozen skin pieces were pulverized under liquid nitrogen using an industrial blender or cryogenic grinder. Soluble proteins were removed by extracting the pig skin powder (0.125 g/ml) with 0.5M sodium acetate overnight at 4° C. The resulting mixture was then centrifuged at 2000 rpm (700×g) at 4° C. for 1 hour. The supernatant was discarded and the extraction procedure repeated three additional times.
  • The resulting pellet was then suspended (0.25 g/ml) in cold MilliQ water and then centrifuged at 2000 rpm (700×g) at 4° C. for 1 hour. The pellet was then washed with water two additional times. Collagen extraction was then performed by suspending the pellet (0.125 g/ml) in 0.075M sodium citrate. The extraction was allowed to proceed for 15-18 hours at 4° C.
  • The resulting mixture was centrifuged at 2000 rpm (700×g) at 4° C. for 1 hour. The supernatant was retained and stored at 4° C. The pellet was re-extracted with 0.075M sodium citrate. The extraction process was repeated such that the tissue was extracted a total of three times. The resulting supernatants were then combined and centrifuged at 9750 rpm (17,000×g) at 4° C. for 1 hour to clarify the solution. The supernatant was retained and the pellet discarded.
  • Collagen was then precipitated from the supernatant by dialyzing (MWCO 12-14,000) extensively against 0.02 M disodium hydrogen phosphate at 4° C. The resulting suspension was then centrifuged at 2000 rpm at 4° C. for 1 hour and the pellet retained. The pellet was then resuspended and rinsed in cold MilliQ water. The suspension was centrifuged at 2000 rpm at 4° C. for 1 hour. The water rinse procedure was repeated two additional times.
  • The resulting collagen pellet was dissolved in 0.1M acetic acid and then lyophilized. The lyophilized material was stored within a dessicator at 4° C. for use in engineering ECMs.
  • EXAMPLE 2 Polyacrylamide Gel Electrophoresis
  • Interrupted gel electrophoresis was performed to compare the protein composition of the collagen preparations made in accordance with the methods described herein to commercially available type I isolated collagen obtained from Sigma-Aldrich, St. Louis, Mo. (Sigma Cat. No. C3511) and pepsin-solubilized collagen from INAMED (Fremont, Calif.) sold under the product name PureCol™. Interrupted gel electrophoresis was performed according to Sykes, et al. The estimation of two collagens from human dermis by interrupted gel electrophoresis. Biochem. Biophys. Res. Commun. 72:1472-1480 (1976), incorporated herein by reference. Sodium dodecylsulfate polyacryamide gel electrophoresis (SDS-PAGE) was performed according to the method of Nielsen, et al. Measurements of molecular weights by gel electrophoresis. Methods in Enzymology, vol. 48, Hirs and Temasheff, Eds., Academic Press, New York, pp. 3-11 (1978), incorporated herein by reference.
  • The evaluation by SDS-PAGE of the protein composition of the collagen preparations made in accordance with the methods described herein indicates that the protocol yielded a pure type I collagen preparation (see below) with minimal to no contaminating non-collagenous proteins (see lanes 2-4 of the gel shown in FIG. 1). The gel shown in FIG. 1 indicates the band pattern of the α1, α2, β11, and β12 chains of collagen. No significant glycosaminoglycan content was identified using a standard alcian blue assay (Bjornsson, S. Simultaneous preparation and quantitation of proteoglycans by precipitation with alcian blue. Anal Biochem. 210:282-291 (1993), incorporated herein by reference).
  • Cyanogen bromide (CNBr) peptide analysis was used to confirm the presence of type I collagen in the pig skin collagen preparation (lanes 2-4), made according to the methods described herein. CNBr peptide analysis was performed according to the method of Miller, et al. Identification of three genetically distinct collagens by cyanogen bromide cleavage of insoluble skin and cartilage collagen. Biochem. Biophys. Res. Commun. 42:1024-1029 (1971). Standard SDS-PAGE within 12% gels was then performed to compare the CNBr-derived peptides from the various collagen preparations (see FIG. 6). The pattern shown in FIG. 6 indicates that the pig skin collagen preparation, made in accordance with the methods described herein, has a number of bands which are similar to Sigma collagen and INAMED collagen. However, there are also bands specific to the pig skin collagen composition. The presence of type I collagen was confirmed in the preparation described herein, and the preparation described herein does not have significant amounts of type V collagen or type III collagen (confirmed using interrupted gel electrophoresis). Types III and V collagen are the other two major types of collagen found in skin.
  • As also shown in FIG. 1, the pig skin type I collagen preparation (lanes 2-4), made according to the methods described herein, yielded a distinct collagen banding pattern compared to the commercially available collagen preparations, namely Sigma Type I collagen (acid solubilized; lane 5) and INAMED collagen (acid/pepsin solubilized; lane 6). Surprisingly, differences between the collagen preparations made in accordance with the methods described herein and the Sigma and INAMED collagen preparations were observed. As noted in FIG. 1 (band designated “Unknown”), a unique high molecular weight protein band was identified within the pig skin collagen preparation made in accordance with the methods described herein. The band was not observed in the INAMED or Sigma commercially available collagen preparations. The unknown protein band was also isolated, digested with CNBr, and a banding pattern indicative of collagen alpha chains was found.
  • EXAMPLE 3 Polymerization Kinetics
  • The polymerization kinetics of the pig skin collagen preparation made in accordance with the methods described herein were also compared to commercially available Sigma Type I collagen. For this evaluation a spectrophotometric turbidity analysis assay for determining polymerization kinetics of collagen, well-known to those skilled in the art was used. The assay was performed in accordance with Comper, et al. Characterization of nuclei in in vitro collagen fibril formation. Biopolymers 16:2133-2142 (1977) and Brightman, et al. Time-lapse confocal reflection microscopy of collagen fibrillogenesis and extracellular matrix assembly in vitro. Biopolymers 54:222-234 (2000), each incorporated herein by reference. The time-course of polymerization was monitored in a Lambda 35 UV-VIS spectrophotometer (Perkin-Elmer) equipped with a temperature-controlled, 8-position cell changer as described previously by Brightman et al., 2000.
  • FIG. 2 shows the time-dependent changes in absorbance as recorded at a wavelength of 405 nm as each of the collagen preparations (each at 2 mg/ml collagen concentration as determined using sirius red analysis) underwent polymerization or fibrillogenesis (i.e., soluble state to an insoluble fibrillar state). The sirius red assay was performed in accordance with Marotta, et al. Sensitive spectrophotometric method for the quantitative estimation of collagen. Anal. Biochem. 150:86-90 (1985), incorporated herein by reference. The pig skin collagen preparation made in accordance with the methods described herein showed a decreased lag time, increased polymerization rate, and increased magnitude of change in A405 readings. Surprisingly, the pig skin collagen preparation made in accordance with the methods described herein exhibited a much decreased lag time and increased polymerization rate in comparison to commercially available Sigma Type I collagen.
  • EXAMPLE 4 Microstructure Comparison Using Confocal Reflection Microscopy
  • The collagen fibril microstructure of a 3D ECM engineered from pig skin collagen, prepared in accordance with the methods described herein, and from commercially available Sigma Type I collagen were determined and compared using confocal reflection microscopy. Each collagen composition was polymerized at 1.5 mg/ml. Confocal Reflection Microscopy was performed according to Brightman, et al. Time-lapse confocal reflection microscopy of collagen fibrillogenesis and extracellular matrix assembly in vitro. Biopolymers 54:222-234 (2000) and Voytik-Harbin, et al. Three-dimensional imaging of extracellular matrix and extracellular matrix-cell interactions. Methods in Cell Biology 63:583-597 (2001), each incorporated herein by reference.
  • Briefly, for example, solutions of collagen were polymerized in a Lab-Tek chambered coverglass and imaged using a BioRad Radiance 2100 MP Rainbow confocal/multiphoton microscope using a 60×1.4 NA oil immersion lens (or an Olympus Fluoview FV 1000 confocal microscope was used). Optical settings were established and optimized for matrices after polymerization was complete. Samples were illuminated with 488 nm laser light and the reflected light detected with a photomultiplier tube (PMT) using a blue reflection filter.
  • The fibril microstructure produced by the pig skin collagen, prepared in accordance with the methods described herein, showed an increase in fibril density and, surprisingly, an apparent increase in fibril-to-fibril association (e.g., cross-linked and/or fibril aggregates; see FIGS. 3A (pig skin collagen preparation)) and 3B (commercially available Sigma Type I collagen)).
  • EXAMPLE 5 Microstructure Comparison Using Scanning Electron Microscopy
  • The collagen fibril microstructure of a 3D ECM engineered from pig skin collagen, prepared in accordance with the methods described herein, and from commercially available Sigma Type I collagen were determined and compared using scanning electron microscopy. Each collagen composition was polymerized at 1.0 mg/ml. The fibril microstructures produced by the pig skin collagen, prepared in accordance with the methods described herein, showed a different appearance than the commercially available Sigma Type I collagen. FIGS. 4A and B show pig skin collagen, prepared in accordance with the methods described herein, and FIGS. 4C and D show commercially available Sigma Type I collagen. Sigma collagen provided a less dense fibril architecture compared to the pig skin collagen. Scanning electron microscopy was performed according to Voytik-Harbin, et al. Small intestinal submucosa: A tissue-derived extracellular matrix that promotes tissue-specific growth and differentiation of cells in vitro. Tissue Engineering, 4:157-174 (1998), incorporated herein by reference. Note that scanning electron microscopy analysis involves fixing and critical point drying and the required processing of the specimens is known to lead to microstructural artifacts. Therefore, this technique alone is not relied on to determine the 3D microstructure of engineered ECMs.
  • EXAMPLE 6 Mechanical Behavior Comparison Using Unconfined Compression
  • FIG. 5 shows the mechanical behavior of a 3D ECM engineered from pig skin collagen (polymerized at 2 mg/ml), prepared in accordance with the methods described herein, and from commercially available Sigma Type I collagen (polymerized at 3 mg/ml). Mechanical behavior was compared using unconfined compression. The specimen was subjected to unconfined compression at a rate of 10 or 20 micrometers/sec using an AR-2000 rheometer (TA Instruments, New Castle, Del.). As shown in FIG. 5, pig skin collagen, prepared in accordance with the methods described herein, surprisingly exhibited as much as 10-20× greater mechanical integrity as commercially available Sigma Type I collagen.
  • EXAMPLE 7 Collagen Polymerization Conditions
  • Briefly, collagen was polymerized by using a 10×PBS, pH 7.4 solution consisting of 1.37M NaCl, 0.027M KCl, 0.081M Na2HPO4, 0.015M KH2PO4, 5 mM MgCl2, and 1% w/v glucose. To polymerize collagen, a mixture was made of 1 ml of solubilized collagen in 0.01N HCl, 150 μl 10×PBS, pH 7.4, 150 μl 0.1N NaOH, 100 μl 13.57 mM CaCl2, and 100 μl 0.01 N HCl. The composition was mixed well after each component was added. Polymerization was allowed to proceed at 37° C.
  • EXAMPLE 8 Shear Modulus During Polymerization
  • Mechanical properties of the engineered ECMs prepared with the pig skin collagen, made according to the method described herein, were measured using a TA Instruments (New Castle, Del.) AR-2000 rheometer. Soluble pig skin and commercially available Sigma collagen preparations were adjusted to provide the polymerization conditions described in Example 7 and were placed on the peltier temperature-controlled lower plate at 4° C., and the 40-mm parallel-plate geometry was lowered to a 1-mm gap. The temperature was then raised to 37° C. to initiate polymerization. The peltier heated plate required about 1 minute to stabilize at 37° C. Measurements of shear storage modulus G′ of the polymerizing material under controlled-strain oscillatory shear were made every 1 minute under oscillation at 1 Hz and 0.02% strain for a specified time.
  • As shown in FIG. 7, for pig skin and Sigma collagen polymerized at the same collagen concentration and using the same polymerization conditions, the shear storage modulus for ECMs prepared from pig skin collagen, according to the methods described herein, is 10 to 40 times greater than that obtained for ECMs prepared from commercially available Sigma collagen.
  • EXAMPLE 9 Stem Cell Growth on Collagen ECM
  • The pig skin collagen preparation, made in accordance with the methods described herein, has been seeded with a number of different cell types, including human mesenchymal stem cells (Clonetics) and a mesenchymal stem cell line derived from the bone marrow of mice (D1, American Type Culture Collection (ATCC)). Experiments have demonstrated that these multi-potential stem cells can be seeded within engineered ECMs prepared from the pig skin collagen and maintain viability. Furthermore, a broader range of fibril microstructures and mechanical properties (stiffness) can be obtained for engineered ECMs prepared from pig skin collagen, according to the methods described herein. The ability to control fibril microstructure and mechanical properties (stiffness) over a broad range is critical to the ability to direct the behavior and differentiation of cells.
  • For example, engineered ECMs were prepared using pig skin collagen, prepared according to the methods described herein, or commercially available Sigma collagen preparations at two concentrations, specifically 0.5 mg/ml and 3 mg/ml. These formulations were used to entrap and culture mouse mesenchymal stem cells within a 3D format. Results showed that engineered ECMs of varied microstructures and mechanical properties (stiffness) could be prepared from pig skin collagen. In turn, mouse mesenchymal stem cells cultured within pig skin ECMs of relatively low stiffness (0.5 mg/ml) showed preferential differentiation down the adipogenic pathway (FIG. 8A) while those prepared with relatively high stiffness (3 mg/ml) supported enhanced osteogenic differentiation (FIG. 8B). Compared to ECMs prepared with commercially available Sigma collagen, those prepared from pig skin supported enhanced osteogenesis. The differences in the two preparations to support osteogenic differentiation of multi-potential stem cells can be attributed to differences in ECM microstructure and mechanical properties. Engineered ECMs of increased mechanical integrity (strength and stiffness) are much more readily obtained using the pig skin collagen preparation, made according to the methods described herein. The cells in FIG. 8 A were cultured for periods of time up to 3 weeks in DMEM supplemented with 10% fetal bovine serum, 0.5 mM isobutyl-methylxanthine, 1 μM dexamethasone, 10 μM insulin, 200 μM indomethacin, 0.3 mg/ml glutamine, 100 U/ml penicillin, and 100 μg/ml streptomycin. The cells in FIG. 8 B were cultured for periods of time up to 3 weeks in DMEM supplemented with 10% fetal bovine serum, 0.3 mg/ml glutamine, 100 U/ml penicillin, and 100 μg/ml streptomycin.
  • EXAMPLE 10 Glycerol Separation of Oligomer and Monomer Components of Collagen Compositions
  • Type I collagen isolated from pig skin was solubilized in acetic acid (0.005 M) to achieve a desired collagen concentration (4 mg/ml). An equal volume of 2× solubilization buffer was added (0.06 M phosphate, 0.2 M NaCl, and 1.2 M glycerol, pH 7.0) with mixing. The solution was left to stand at 30° C. for 7 days. At desired times, the solution was then centrifuged at 10,000 rpm and 4° C. for 10 minutes and decanted and the supernatant was retained. The pellet was then resolubilized in 0.1 M acetic acid and the resolubilized pellet and the supernatant were dialyzed against 0.01 M acetic acid and lyophilized to dryness.
  • EXAMPLE 11 Analysis of Collagen Fibril Microstructure
  • Confocal reflection (CRM) microscopy was used to collect high resolution images of ECMs (without cells). Fibril density and fibril diameters were measured from the images to quantify the effect of HA concentration on the collagen fibrillar microstructure. Confocal imaging was performed on a Olympus Fluoview FV1000 confocal system adapted to a IX81 inverted microscope (Olympus, Tokyo, Japan). For the fibril density, confocal image stacks were collected from random locations within independent ECMs for each treatment. The fibril density for each image was calculated in Matlab (Mathworks, Natick, Mass.) as the ratio of fibril volume (voxels containing fibrils) to total volume after binarizing (thresholding) the images. Fibril diameters were measured from the 2D projections of the confocal images in Imaris 5.0 (Bitplane Inc., Saint Paul, Minn.). Each fibril diameter represents the average of 5 diameters measured along the major axis of an individual fibril.
  • EXAMPLE 12 Rheometry Protocol
  • About 1.5 ml of collagen was neutralized according to the Sirius red concentration in a 2 ml eppendorf tube (all reagents were on ice). A balance was used to acquire the proper amount of collagen assuming 1 g=1 ml (a weight within 3 mg is acceptable). The weight was recorded to the nearest mg. Proper amounts of HCL and PBS were added and the tube was shaken moderately. NaOH was added and the tube was shaken moderately. Finally, CaCl and HCL were added and the tube was shaken. The tubes were then centrifuged 10K. The sample was removed and 1 ml of the sample was added to a cold rheometer plate (no bubbles should be allowed into the solution on the plate and excessive condensation should not be allowed on the cold plate). The geometry was spun lightly and the geometry was lowered to a 725 μm gap position (this position correlates to 1 ml). The spinning motion helped to insure that the sample was evenly distributed under the geometry. Once the geometry was at the proper gap, it was verified that the collagen solution extended to the edge of the plate, but not past it. The vapor trap was placed on the plate, and the normal force was zeroed. A reading was then taken.
  • EXAMPLE 13 Glycerol Separation of Oligomers and Monomers
  • The glycerol separation was preformed according to the method described in Example 10. FIG. 9 shows the time-dependent glycerol-based separation of pig skin collagen type I oligomers and monomers. At each time point (0, 6, 24, 48, and 72 hours), the supernatant and pellet represent the monomer-rich and oligomer-rich fractions, respectively. By definition collagen monomers represent single collagen molecules and oligomers represent covalently cross-linked monomers (e.g., dimers=2 monomers, trimers=3 monomers, etc.). The results in FIG. 9 show that the pellet is enriched in the higher molecular weight oligomeric collagen molecules.
  • EXAMPLE 14 Densitometry Readings after Glycerol-Based Separation of Oligomers and Monomers
  • FIG. 10A shows glycerol-based separation of pig skin collagen type I oligomers and monomers. Pig skin collagen was polymerized in the presence of glycerol for 7 days as described in Example 10. The mixture was centrifuged and the supernatant and pellet retained, dialyzed against dilute acetic acid, and lyophilized to dryness. The supernatant and pellet represented monomer- and oligomer-rich collagen compositions, respectively, as determined by SDS-PAGE (4%). FIG. 10A shows the SDS-PAGE gel of the samples and FIG. 10B shows the corresponding densitometry readings. With reference to the labeling of bands shown in FIG. 9, the densitometry readings show a comparison of the oligomer-rich and monomer-rich fraction densitometry readings for individual bands on the SDS-PAGE gel relative to densitometry readings for alpha 1. For the densitometry results, the leftmost bar for each group of 3 bars is the initial (before glycerol separation) densitometry reading for each band relative to alpha 1 in the initial isolated collagen preparation, the middle bar for each group of 3 bars is the desitometry reading for each band in the oligomer-rich fraction after glycerol-based separation relative to alpha 1, and the rightmost bar for each group of 3 bars is the desitometry reading for each band in the monomer-rich fraction after glycerol-based separation relative to alpha 1. The results show that for known oligomers (e.g., β12), the oligomers are found mainly in the pellet after glycerol-based separation. SDS-PAGE analysis was performed as described in Example 2.
  • EXAMPLE 15 CNBr Peptide Analysis of Collagen Oligomers
  • FIG. 11 shows SDS-PAGE (12%) analysis of CNBr peptide maps of collagen preps in which oligomer:monomer content is varied. Upon CNBr digestion, collagen preps yielded distinct peptide fingerprints indicating distinct molecular compositions and cross-linked moieties. SDS-PAGE analysis and CNBr peptide analysis were performed as described in Example 3.
  • EXAMPLE 16 Polymerization Kinetics of Oligomeric Collagen
  • Polymerization kinetics were measured as described in Example 3. FIGS. 12 A-B show the oligomer:monomer content of pig skin collagen preparation affects polymerization (self-assembly) kinetics. An increase in oligomer content decreases lag time (FIG. 12 B) and increases the rate of polymerization (FIG. 12 A). FIG. 12A shows A405 vs time. In FIG. 12 A, the uppermost line is the control (0.7 mg/ml collagen) which is the resuspended collagen pellet after collagen isolation, but prior to glycerol separation. The lowest line is 100% monomer-rich collagen. The middle lines from top to bottom are 1.) 5.4% monomer-rich collagen and 94.6% oligomer-rich collagen, 2.) 100% oligomer-rich collagen, and 3.) 55.4% monomer rich+44.6% oligomer-rich collagen.
  • EXAMPLE 17 Mechanical Behavior of Extracellular Matrices Prepared with Oligomeric Collagen
  • FIG. 13 shows the results of an assay done to compare mechanical behavior of 3D engineered matrices prepared with pig skin collagen preps of varied monomer:oligomer content. The shear storage modulus (G′) provides a measure of the stiffness of the solid (elastic) components. The results in FIG. 13 show that the shear storage modulus (G′; stiffness) of matrices increases with oligomer content. The top line is the control. The bottom line is 100% monomer-rich collagen. The middle lines are (from top to bottom) 5.4% monomer-rich collagen and 94.6% oligomer-rich collagen, 100% oligomer-rich collagen, and 55.4% monomer rich+44.6% oligomer-rich collagen.
  • EXAMPLE 18 Mechanical Behavior of Extracellular Matrices Prepared with Oligomeric Collagen
  • FIG. 14 shows the comparison of mechanical behavior of 3D engineered matrices prepared with pig skin collagen preps of varied monomer:oligomer content. An increase in delta as a function of strain as observed with the monomer-rich preparation (uppermost line) indicates more fluid-like (viscous) behavior.
  • EXAMPLE 19 Mechanical Behavior of Extracellular Matrices Prepared with Oligomeric Collagen
  • FIG. 15 shows a comparison of mechanical behavior of 3D engineered matrices prepared with pig skin collagen preps of varied monomer:oligomer content. Matrices produced with collagen preps consisting of higher oligomer content show increased compressive stiffness. The bottom line is monomer-rich collagen. The next line towards the top is the control. The top three lines are, from top to bottom, 100% oligomer-rich collagen, 5.4% monomer-rich collagen and 94.6% oligomer-rich collagen, and 55.4% monomer rich+44.6% oligomer-rich collagen.
  • EXAMPLE 20 Mechanical Behavior of Extracellular Matrices Prepared with Oligomeric Collagen
  • FIG. 16 shows the results of an assay done to determine the compressive stiffness values as a function of the oligomer:monomer content of the pig skin collagen preparation. The results show that stiffness increases with increasing oligomer content.
  • EXAMPLE 21 Confocal Microscopy to Visualize Engineered Matrices Made with Varying Oligomer Content
  • FIG. 17 shows the effects of oligomer content on the 3D collagen fibril microstructure of engineered matrices. The matrices were prepared with the percentages of oligomers and monomers shown. The oligomer-rich and monomer-rich preparations were 100% oligomer-rich and 100% monomer-rich, respectively. Confocal microscopy was performed as described in Example 4.
  • EXAMPLE 22 Confocal Microscopy to Visualize Engineered Matrices Made with Varying Oligomer Content
  • FIG. 18 shows the effects of oligomer content on the 3D collagen fibril microstructure of engineered matrices. The matrices were prepared with the percentages of oligomers and monomers shown. The oligomer-rich and monomer-rich preparations were 100% oligomer-rich and 100% monomer-rich, respectively. Confocal microscopy was performed as described in Example 4.
  • EXAMPLE 23 SDS-PAGE Analysis of Collagen Preparations
  • FIG. 19 shows the compositional analysis of different collagen preparations as determined using SDS-PAGE (4%). The results in FIG. 19 show that the pig skin collagen preparation uniquely contains collagen oligomers that run intermediate between commonly identified dimeric (β) and trimeric (γ) forms of collagen. Upon pepsin treatment of pig skin collagen, the content of unique collagen oligomers as well as other higher molecular weight collagen species was significantly reduced. Lane 1=PS+pepsin (acid solubilized type I collagen from pig skin posted-treated with pepsin); Lane 2=Vitrogen (pepsin solubilized type I collagen from calf skin); Lane 3=PureCol (pepsin solubilized type I collagen from calf skin); Lane 4=(Sigma, A) acid solubilized type I collagen from calf skin, Lot A; Lane 5=(Sigma, B) acid solubilized type I collagen from calf skin, Lot B; Lane 6=PS (acid solubilized type I collagen from pig skin). SDS-PAGE analysis and CNBr peptide analysis were performed as described in Example 3.
  • EXAMPLE 24 Polymerization Kinetics of Different Collagen Preparations
  • FIG. 20 shows the kinetics of polymerization for the different collagen preparations as determined using the spectrophotometric-based turbidity assay described in Example 3. Results show that the polymerization kinetics of collagen is highly dependent upon the collagen preparation. More specifically, polymerization of pig skin collagen shows a decreased lag phase and an increased rate of polymerization (slope of growth phase). Pig skin collagen (0.25 mg/ml) and pig skin collagen (1.0 mg/ml) are the lower and upper left-most lines, respectively. Sigma collagen (0.25 mg/ml) and Sigma collagen (1.0 mg/ml) are the lowest line and the line where the third highest absorbance was achieved, respectively. PureCol (0.25 mg/ml) and PureCol (1.0 mg/ml) are the line with the greatest lag time and the line where the highest absorbance was achieved, respectively.
  • EXAMPLE 25 Polymerization of Different Collagen Preparations
  • FIG. 21 shows a comparison of final absorbance (405 nm) values for different collagen preparations as determined from a spectrophometric based turbidity assay as described in Example 3. Results represent average of the last 10 absorbance values.
  • EXAMPLE 26 Confocal Microscopy to Visualize Different Engineered Matrices
  • FIG. 22 shows the collagen fibril microstructure of 3D engineered ECMs prepared with different purified type I collagen sources. Each collagen preparation was polymerized at two different concentrations (0.5 mg/ml and 2 mg/ml) using the same reaction conditions. Images represent 2D projections of 3D image stacks as collected using confocal reflection microscopy as described in Example 4. The results show that the collagen fibril microstructure of 3D engineered ECMs is highly dependent upon the collagen preparation.
  • EXAMPLE 27 Mechanical Behavior of Extracellular Matrices Prepared with Different Collagen Preparations
  • FIG. 23 shows the comparison of mechanical behavior of 3D engineered ECMs prepared with different collagen preparations and tested in oscillatory shear. The storage modulus (G′) provides a measure of the stiffness of the solid (elastic) components. Results show that the stiffness of matrices prepared with pig skin collagen are significantly greater than those obtained with the commercial collagen preparations (PureCol and Sigma).
  • EXAMPLE 28 Mechanical Behavior of Extracellular Matrices Prepared with Different Collagen Preparations
  • FIGS. 24A-B show a comparison of mechanical behaviors of engineered matrices prepared with different collagen sources. FIG. 24 A shows microstructural analysis of engineered ECMs using confocal reflection microscopy (fibril density vs. collagen concentration). FIG. 24 B shows microstructural analysis of engineered ECMs using confocal reflection microscopy (average fibril diameter vs. collagen concentration). Collagen stiffness (G′ vs. fibril density) and rheometric analysis of mechanical properties (G′ (Pa) vs. collagen concentration) were also performed. Confocal microscopy was as described in Example 4.
  • EXAMPLE 29 Effect of Oligomer Content on Polymerization Efficiency
  • Table 1 shows the effect of oligomer content on polymerization efficiency. Polymerization was assayed as described in Example 4. The collagen that remained in solution as polymerized versus non-polymerized collagen was determined based on a Sirius Red assay. The results in table 1 show that oligomer-rich collagen polymerizes more efficiently than monomer-rich collagen.
  • TABLE 1
    Effect of Oligomer Content on Polymerization Efficiency.
    Collagen Collagen Concentration % Polymerized (t = 1
    Preparation (mg/mL) hour)
    Initial (Control) 0.82  100%
    0.7  100%
    0.21  100%
    Oligomer-Rich 0.7  100%
    0.57  100%
    0.14  100%
    Monomeric-Rich 0.7 86.6%
    0.36 81.4%
    0.09 58.4%
  • EXAMPLE 30 Half-Polymerization Times for Different Collagen Preparations
  • Table 1 shows the t ½'s for polymerization for different collagen preparations. Polymerization was assayed as described in Example 4. The results in Table 2 show that oligomer-rich pig skin collagen polymerizes with a t ½ that is much lower than Sigma collagen or PureCol. A t ½ is the half-time required to achieve maximal absorbance.
  • TABLE 2
    Comparison of half-polymerization times (t1/2) for different collagen
    preparations as determined from spectrophometric based
    turbidity assay.
    Collagen Preparation t1/2 (minutes)
    PSC (Acid Solubilized Type I Collagen from Pig Skin)  1-1.5
    PureCol (Pepsin Solubilized Type I Collagen from Calf 13-17
    Skin)
    Sigma (Acid Solubilized Type I Collagen from Calf Skin)  5-9

Claims (25)

1. A method for isolating collagen, said method comprising the steps of:
obtaining a collagen-containing source material;
comminuting the source material;
mixing the comminuted source material with an extraction solution;
extracting the comminuted source material to form a soluble fraction and an insoluble fraction;
obtaining the insoluble fraction;
extracting the collagen from the insoluble fraction to form a soluble collagen fraction;
precipitating the collagen from the soluble collagen fraction; and
resuspending the precipitate in an aqueous solution wherein the aqueous solution used to resuspend the collagen precipitate is an acidic solution.
2. The method of claim 1 wherein the collagen-containing source material is porcine skin tissue.
3. The method of claim 1 wherein the source material is frozen in liquid nitrogen prior to, during, or prior to and during the comminuting step.
4. The method of claim 1 further comprising the step of lyophilizing the collagen precipitate.
5. The method of claim 1 wherein the collagen is precipitated by dialysis against a buffered solution.
6. The method of claim 5 wherein the dialysis tubing has a molecular weight cut-off of about 12,000 to about 14,000.
7. A method for engineering matrices with enhanced polymerization characteristics, said method comprising the steps of
obtaining collagen oligomers;
polymerizing the collagen oligomers;
and forming the engineered matrices with enhanced polymerization characteristics.
8. The method of claim 7 wherein the enhanced polymerization characteristics are selected from the group consisting of an enhanced rate of polymerization, a reduced lag time for polymerization, and an enhanced mechanical integrity.
9. The method of claim 7 wherein the enhanced mechanical integrity is selected from the group consisting of strength and stiffness.
10. The method of claim 7 wherein the collagen oligomers polymerize with a maximal t ½ selected from the group consisting of about 3 minutes, about 2.5 minutes, about 2.0 minutes, about 1.5 minutes, and about 1 minute.
11. The method of claim 7 wherein the collagen oligomers are obtained by isolation from a collagen-containing source material that is a tissue naturally enriched with collagen oligomers.
12. The method of claim 7 wherein the collagen oligomers are obtained by isolating and then chemically cross-linking collagen.
13. The method of claim 7 wherein the collagen oligomers are isolated from a diseased tissue or a genetically-modified tissue.
14. An engineered matrix comprising a predetermined percentage of collagen oligomers based on total isolated collagen used to make the engineered matrix.
15. The matrix of claim 14 wherein the collagen is isolated from a tissue naturally enriched with collagen oligomers.
16. The matrix of claim 14 wherein the collagen oligomers are isolated from a diseased tissue or a genetically-modified tissue.
17. The matrix of claim 14 wherein the collagen oligomers are obtained by isolating and then chemically cross-linking collagen.
18. The matrix of claim 14 wherein the predetermined percentage of collagen oligomers is selected from the group consisting of about 0.5% to about 100%, about 1.0% to about 100%, about 2% to about 100%, about 3% to about 100%, about 5% to about 100%, about 10% to about 100%, about 15% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 90% to about 100%, about 95% to about 100%, and about 100%.
19. The matrix of claim 14 wherein the engineered matrix further comprises a predetermined percentage of isolated collagen monomers.
20. A graft composition comprising an engineered matrix comprising collagen oligomers wherein the matrix has a predetermined percentage of collagen oligomers based on total isolated collagen used to make the engineered matrix.
21. The graft composition of claim 20 wherein the predetermined percentage of collagen oligomers is selected from the group consisting of about 0.5% to about 100%, about 1.0% to about 100%, about 2% to about 100%, about 3% to about 100%, about 5% to about 100%, about 10% to about 100%, about 15% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 90% to about 100%, about 95% to about 100%, and about 100%.
22. The graft composition of claim 20 wherein the collagen oligomers are obtained from a natural collagen-containing source material enriched with collagen oligomers.
23. The graft composition of claim 20 wherein the collagen oligomers are isolated from a diseased tissue or a genetically-modified tissue.
24. The graft composition of claim 20 wherein the collagen oligomers are obtained by isolating and then chemically cross-linking collagen.
25. The graft composition of claim 20 wherein the engineered matrix further comprises a predetermined percentage of collagen monomers based on total isolated collagen used to make the engineered matrix.
US11/903,326 2006-09-21 2007-09-21 Collagen preparation and method of isolation Active US8084055B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/903,326 US8084055B2 (en) 2006-09-21 2007-09-21 Collagen preparation and method of isolation
US13/331,483 US8512756B2 (en) 2006-09-21 2011-12-20 Collagen preparation and method of isolation

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US84620706P 2006-09-21 2006-09-21
US11/903,326 US8084055B2 (en) 2006-09-21 2007-09-21 Collagen preparation and method of isolation

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/331,483 Continuation US8512756B2 (en) 2006-09-21 2011-12-20 Collagen preparation and method of isolation

Publications (2)

Publication Number Publication Date
US20080268052A1 true US20080268052A1 (en) 2008-10-30
US8084055B2 US8084055B2 (en) 2011-12-27

Family

ID=38988112

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/903,326 Active US8084055B2 (en) 2006-09-21 2007-09-21 Collagen preparation and method of isolation
US13/331,483 Active US8512756B2 (en) 2006-09-21 2011-12-20 Collagen preparation and method of isolation

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/331,483 Active US8512756B2 (en) 2006-09-21 2011-12-20 Collagen preparation and method of isolation

Country Status (5)

Country Link
US (2) US8084055B2 (en)
AU (1) AU2007297611B2 (en)
CA (1) CA2663722C (en)
GB (1) GB2455041B (en)
WO (1) WO2008036393A1 (en)

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070269476A1 (en) * 2006-05-16 2007-11-22 Voytik-Harbin Sherry L Engineered extracellular matrices control stem cell behavior
US20090280180A1 (en) * 2007-12-10 2009-11-12 Voytik-Harbin Sherry L Collagen-based matrices with stem cells
US20110021753A1 (en) * 2009-07-27 2011-01-27 National Cheng Kung University Method for Preparing Porous Collagen Matrices
US20110020216A1 (en) * 2007-06-21 2011-01-27 David James Mooney Scaffolds for cell collection or elimination
US20110020864A1 (en) * 2009-07-27 2011-01-27 National Cheng Kung University Preparation of High Purity Collagen
US20110117170A1 (en) * 2008-05-30 2011-05-19 Lan Cao Controlled Release of Growth Factors and Signaling Molecules for Promoting Angiogenesis
US8084055B2 (en) 2006-09-21 2011-12-27 Purdue Research Foundation Collagen preparation and method of isolation
US20120027732A1 (en) * 2010-07-27 2012-02-02 Voytik-Harbin Sherry L Thermoreversible collagen
US20120077181A1 (en) * 2009-02-19 2012-03-29 Timo Schmidt Method for the cryopreservation of cells, artificial cell constructs or three-dimensional complex tissues assemblies
US20120122218A1 (en) * 2009-04-13 2012-05-17 President And Fellows Of Harvard College Harnessing Cell Dynamics to Engineer Materials
WO2013013537A1 (en) 2011-07-28 2013-01-31 Wang Shanshan Composite collagen sponge and preparation method thereof
US8932583B2 (en) 2005-12-13 2015-01-13 President And Fellows Of Harvard College Scaffolds for cell transplantation
US9370558B2 (en) 2008-02-13 2016-06-21 President And Fellows Of Harvard College Controlled delivery of TLR agonists in structural polymeric devices
US9381235B2 (en) 2009-07-31 2016-07-05 President And Fellows Of Harvard College Programming of cells for tolerogenic therapies
US9486512B2 (en) 2011-06-03 2016-11-08 President And Fellows Of Harvard College In situ antigen-generating cancer vaccine
US9603894B2 (en) 2010-11-08 2017-03-28 President And Fellows Of Harvard College Materials presenting notch signaling molecules to control cell behavior
US9610328B2 (en) 2010-03-05 2017-04-04 President And Fellows Of Harvard College Enhancement of skeletal muscle stem cell engraftment by dual delivery of VEGF and IGF-1
US9675561B2 (en) 2011-04-28 2017-06-13 President And Fellows Of Harvard College Injectable cryogel vaccine devices and methods of use thereof
US9693954B2 (en) 2010-06-25 2017-07-04 President And Fellows Of Harvard College Co-delivery of stimulatory and inhibitory factors to create temporally stable and spatially restricted zones
US9821045B2 (en) 2008-02-13 2017-11-21 President And Fellows Of Harvard College Controlled delivery of TLR3 agonists in structural polymeric devices
US9878071B2 (en) 2013-10-16 2018-01-30 Purdue Research Foundation Collagen compositions and methods of use
US9937249B2 (en) 2012-04-16 2018-04-10 President And Fellows Of Harvard College Mesoporous silica compositions for modulating immune responses
WO2018144496A1 (en) * 2017-01-31 2018-08-09 Geniphys, Llc Methods and compositions for matrix preparation
US10045947B2 (en) 2011-04-28 2018-08-14 President And Fellows Of Harvard College Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
US10647959B2 (en) 2011-04-27 2020-05-12 President And Fellows Of Harvard College Cell-friendly inverse opal hydrogels for cell encapsulation, drug and protein delivery, and functional nanoparticle encapsulation
US10682400B2 (en) 2014-04-30 2020-06-16 President And Fellows Of Harvard College Combination vaccine devices and methods of killing cancer cells
US10828337B2 (en) 2011-06-02 2020-11-10 Indiana University Research And Technology Corporation Materials and methods for controlling vasculogenesis from endothelial colony forming cells
US11150242B2 (en) 2015-04-10 2021-10-19 President And Fellows Of Harvard College Immune cell trapping devices and methods for making and using the same
US11202759B2 (en) 2010-10-06 2021-12-21 President And Fellows Of Harvard College Injectable, pore-forming hydrogels for materials-based cell therapies
CN115466322A (en) * 2022-09-26 2022-12-13 斐缦(长春)医药生物科技有限责任公司 Self-assembled collagen and preparation method and application thereof
US11555177B2 (en) 2016-07-13 2023-01-17 President And Fellows Of Harvard College Antigen-presenting cell-mimetic scaffolds and methods for making and using the same
US11590256B2 (en) * 2017-11-03 2023-02-28 Cellontech Co., Ltd. Medical material produced using collagen and method for producing same
US11739291B2 (en) 2017-04-25 2023-08-29 Purdue Research Foundation 3-dimensional (3D) tissue-engineered muscle for tissue restoration
US11752238B2 (en) 2016-02-06 2023-09-12 President And Fellows Of Harvard College Recapitulating the hematopoietic niche to reconstitute immunity
US11786457B2 (en) 2015-01-30 2023-10-17 President And Fellows Of Harvard College Peritumoral and intratumoral materials for cancer therapy
US11919941B2 (en) 2016-04-21 2024-03-05 Purdue Research Foundation Cell-collagen-silica composites and methods of making and using the same

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2441098B (en) * 2005-05-16 2010-05-26 Purdue Research Foundation Engineered extracellular matrices
CA2640268A1 (en) 2006-01-25 2007-08-02 Children's Medical Center Corporation Methods and procedures for ligament repair
CA2664866C (en) 2006-09-28 2018-08-14 Children's Medical Center Coporation Methods and collagen products for tissue repair
EP2566567A4 (en) * 2010-05-07 2014-11-26 Univ North Carolina Method of engrafting cells from solid tissues
KR101446265B1 (en) * 2011-10-17 2014-11-03 메디칸(주) Bio-Fat material eliminated immunity
AU2013214827A1 (en) 2012-02-01 2014-08-21 Children's Medical Center Corporation Biomaterial for articular cartilage maintenance and treatment of arthritis
EP3798226A1 (en) * 2013-02-01 2021-03-31 Children's Medical Center Corporation Collagen scaffolds
CN105209005B (en) 2013-03-04 2019-03-12 德梅尔有限责任公司以埃特诺根有限责任公司名义经营 Injectable can in-situ polymerization collagen compositions
JP2017529390A (en) * 2014-08-27 2017-10-05 パデュー リサーチ ファウンデーション オフィス オブ テクノロジー コマーシャリゼーション Collagen-based therapeutic delivery system
WO2016172365A1 (en) * 2015-04-21 2016-10-27 Purdue Research Foundation Office Of Technology Commercialization Cell-collagen-silica composites and methods of making and using the same
CN110678590B (en) * 2017-05-31 2022-07-19 爱德华兹生命科学公司 Collagen fibers and articles formed therefrom
US20220026435A1 (en) * 2018-11-14 2022-01-27 Avicenna Nutraceutical, Llc Kits and methods for quantifying collagen

Citations (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3949073A (en) * 1974-11-18 1976-04-06 The Board Of Trustees Of Leland Stanford Junior University Process for augmenting connective mammalian tissue with in situ polymerizable native collagen solution
US4233360A (en) * 1975-10-22 1980-11-11 Collagen Corporation Non-antigenic collagen and articles of manufacture
US4544516A (en) * 1982-07-28 1985-10-01 Battelle Development Corporation Collagen orientation
US4582640A (en) * 1982-03-08 1986-04-15 Collagen Corporation Injectable cross-linked collagen implant material
US4789663A (en) * 1984-07-06 1988-12-06 Collagen Corporation Methods of bone repair using collagen
US4902508A (en) * 1988-07-11 1990-02-20 Purdue Research Foundation Tissue graft composition
US4956178A (en) * 1988-07-11 1990-09-11 Purdue Research Foundation Tissue graft composition
US5163955A (en) * 1991-01-24 1992-11-17 Autogenics Rapid assembly, concentric mating stent, tissue heart valve with enhanced clamping and tissue alignment
US5204382A (en) * 1992-02-28 1993-04-20 Collagen Corporation Injectable ceramic compositions and methods for their preparation and use
US5275826A (en) * 1992-11-13 1994-01-04 Purdue Research Foundation Fluidized intestinal submucosa and its use as an injectable tissue graft
US5281422A (en) * 1991-09-24 1994-01-25 Purdue Research Foundation Graft for promoting autogenous tissue growth
US5420248A (en) * 1991-07-04 1995-05-30 Coletica Unpigmented fish skin, particularly from flat fish, as a novel industrial source of collagen, extraction method, collagen and biomaterial thereby obtained
US5460962A (en) * 1994-01-04 1995-10-24 Organogenesis Inc. Peracetic acid sterilization of collagen or collagenous tissue
US5554389A (en) * 1995-04-07 1996-09-10 Purdue Research Foundation Urinary bladder submucosa derived tissue graft
US5885619A (en) * 1995-04-07 1999-03-23 Purdue Research Foundation Large area submucosal tissue graft constructs and method for making the same
US5948429A (en) * 1994-11-22 1999-09-07 Tissue Engineering, Inc. Methods for preparing biopolymer foams
US6099567A (en) * 1996-12-10 2000-08-08 Purdue Research Foundation Stomach submucosa derived tissue graft
US6187047B1 (en) * 1995-10-16 2001-02-13 Orquest, Inc. Bone grafting matrix
US6206931B1 (en) * 1996-08-23 2001-03-27 Cook Incorporated Graft prosthesis materials
US6264992B1 (en) * 1998-02-27 2001-07-24 Purdue Research Foundation Submucosa as a growth substrate for cells
US6375989B1 (en) * 1996-12-10 2002-04-23 Purdue Research Foundation Submucosa extracts
US6384196B1 (en) * 1998-03-24 2002-05-07 Merck Patent Gesellschaft Process for the preparation of mineralized collagen fibrils and their uses as bone substitute material
US20020076816A1 (en) * 2000-12-04 2002-06-20 Jianwu Dai Stem cells and signals developed for use in tissue and organ repair and replacement
US20020170120A1 (en) * 1999-12-07 2002-11-21 Zdenek Eckmayer Method of making a collagen membrane from porcine skin
US6497875B1 (en) * 1996-04-26 2002-12-24 Case Western Reserve University Multilayer skin or dermal equivalent having a layer containing mesenchymal stem cells
US6576265B1 (en) * 1999-12-22 2003-06-10 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US6592794B1 (en) * 1999-09-28 2003-07-15 Organogenesis Inc. Process of making bioengineered collagen fibrils
US6592623B1 (en) * 1999-08-31 2003-07-15 Virginia Commonwealth University Intellectual Property Foundation Engineered muscle
US6666892B2 (en) * 1996-08-23 2003-12-23 Cook Biotech Incorporated Multi-formed collagenous biomaterial medical device
US20040037813A1 (en) * 1999-02-25 2004-02-26 Simpson David G. Electroprocessed collagen and tissue engineering
US20040137616A1 (en) * 2000-02-29 2004-07-15 Isseroff Roslyn R. Corneal epithelial graft composites
US6793939B2 (en) * 1996-12-10 2004-09-21 Purdue Research Foundation Biomaterial derived from vertebrate liver tissue
US20050014181A1 (en) * 2003-06-18 2005-01-20 Galis Zorina S. Methods and compositions for the assessment of polymer assembly
US20050019419A1 (en) * 2002-01-11 2005-01-27 Badylak Stephen Francis Biomaterial derived from vertebrate liver tissue
US6893812B2 (en) * 2000-05-30 2005-05-17 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Three-dimensional ex vivo angiogenesis system
US20050153442A1 (en) * 1999-03-10 2005-07-14 Adam Katz Adipose-derived stem cells and lattices
US20050226856A1 (en) * 2004-03-09 2005-10-13 Ahlfors Jan-Eric W Autogenic living scaffolds and living tissue matrices: methods and uses thereof
US20050260748A1 (en) * 2004-02-27 2005-11-24 Michigan State University Adult stem cells and uses thereof
US20050266556A1 (en) * 2004-02-09 2005-12-01 Yoder Mervin C Isolation, expansion and use of clonogenic endothelial progenitor cells
US20060147501A1 (en) * 2003-02-28 2006-07-06 Hillas Patrick J Collagen compositions and biomaterials
US20060165667A1 (en) * 2004-12-03 2006-07-27 Case Western Reserve University Novel methods, compositions and devices for inducing neovascularization
US20060235511A1 (en) * 2004-02-09 2006-10-19 Cook Incorporated Woven implantable device
US20070026518A1 (en) * 2005-03-29 2007-02-01 The Regents Of The University Of California Controlling stem cell destiny with tunable matrices
US20070077652A1 (en) * 2004-09-16 2007-04-05 Tony Peled Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells
US20070269476A1 (en) * 2006-05-16 2007-11-22 Voytik-Harbin Sherry L Engineered extracellular matrices control stem cell behavior
US20080199441A1 (en) * 2006-11-09 2008-08-21 Tony Peled Use of ex-vivo cultured hematopoietic cells for treatment of peripheral vascular diseases
US20090011021A1 (en) * 2005-05-16 2009-01-08 Purdue Research Foundation Engineered Extracellular Matrices
US20090069893A1 (en) * 2007-04-19 2009-03-12 Mikhail Vitoldovich Paukshto Oriented Collagen-Based Materials, Films and Methods of Making Same
US20090175922A1 (en) * 2006-05-16 2009-07-09 Voytik-Harbin Sherry L Three dimensional purified collagen matrices
US20090280180A1 (en) * 2007-12-10 2009-11-12 Voytik-Harbin Sherry L Collagen-based matrices with stem cells

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4600533A (en) 1984-12-24 1986-07-15 Collagen Corporation Collagen membranes for medical use
JPH0774239B2 (en) * 1986-09-04 1995-08-09 新田ゼラチン株式会社 Manufacturing method of high gel strength acid soluble collagen for cell culture
US5067961A (en) 1988-02-18 1991-11-26 Autogenesis Technologies, Inc. Non-biodegradable two phase corneal implant and method for preparing same
US5428022A (en) 1992-07-29 1995-06-27 Collagen Corporation Composition of low type III content human placental collagen
US5714582A (en) * 1995-03-17 1998-02-03 Bioscience Consultants Invertebrate type V telopeptide collagen, methods of making, and use thereof
ES2249928T3 (en) 1998-11-19 2006-04-01 Organogenesis Inc. MANIPULATED FABRIC CONSTRUCTS THROUGH BIOINGENIERIA AND METHODS TO PRODUCE AND USE THEM.
ES2325977T3 (en) 1999-09-28 2009-09-28 Organogenesis Inc. COLLAGEN FIBRILLES OBTAINED BY BIOTECHNOLOGY.
DE60031461T2 (en) 1999-12-22 2007-08-23 Acell, Inc., Cambridge Composition for tissue regeneration
CZ301649B6 (en) 2000-06-28 2010-05-12 Ed. Geistlich Soehne Ag Fur Chemische Industrie Incorporated Under The Laws Of Switzerland Tube for regeneration of nerves and process for producing thereof
US7029689B2 (en) 2001-05-10 2006-04-18 Georgia Tech Research Corporation Tubular construct for implantation
CA2388723C (en) 2001-06-06 2012-10-23 Becton, Dickinson & Company Method of providing a substrate with a ready-to-use, uniformly distributed extracellular matrix
WO2002102237A2 (en) 2001-06-15 2002-12-27 The Cleveland Clinic Foundation Tissue engineered mitral valve chrodae and methods of making and using same
EP1270672B1 (en) * 2001-06-28 2007-11-14 PV Industries B.V. Process for recovering native collagen
DE20115753U1 (en) 2001-09-14 2002-01-10 Inst Agrar Und Stadtoekologisc Soluble collagens
US8394371B2 (en) 2002-02-11 2013-03-12 Neocutis Sa Compositions for the treatment of skin conditions, disorders or diseases and methods of making and using the same
EP1499181A4 (en) 2002-04-12 2007-11-14 Univ Yale Vascularized human skin equivalent
AUPS242702A0 (en) * 2002-05-21 2002-06-13 Colltech Australia Limited Improved method for the extraction and purification of collagen
US20060258560A1 (en) 2002-09-30 2006-11-16 Chunlin Yang Dry tissue sealant compositions
US20040126405A1 (en) 2002-12-30 2004-07-01 Scimed Life Systems, Inc. Engineered scaffolds for promoting growth of cells
GB0415080D0 (en) 2004-07-05 2004-08-04 Ucl Biomedica Plc Methods for preparing tissue equivalent implants and products thereof
US20060134072A1 (en) * 2004-12-22 2006-06-22 Pedrozo Hugo A Self-assembling protein matrix prepared from natural extracellular matrices
WO2006125025A2 (en) 2005-05-16 2006-11-23 Purdue Research Foundation Engineered extracellular matrices control stem cell behavior
US20100068804A1 (en) 2005-09-01 2010-03-18 Duke University Methods of stimulating expansion of hematopoietic stem cells
US8084055B2 (en) 2006-09-21 2011-12-27 Purdue Research Foundation Collagen preparation and method of isolation
MX354253B (en) 2006-10-06 2018-02-20 Anthrogenesis Corp Human placental collagen compositions, and methods of making and using the same.
WO2010123928A1 (en) 2009-04-20 2010-10-28 Purdue Research Foundation Collagen-based matrices for vessel density and size regulation
US20120115222A1 (en) * 2009-07-16 2012-05-10 Purdue Research Foundation Composition and method for maintenance, differentiation, and proliferation of stem cells
US20120027732A1 (en) 2010-07-27 2012-02-02 Voytik-Harbin Sherry L Thermoreversible collagen

Patent Citations (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3949073A (en) * 1974-11-18 1976-04-06 The Board Of Trustees Of Leland Stanford Junior University Process for augmenting connective mammalian tissue with in situ polymerizable native collagen solution
US4233360A (en) * 1975-10-22 1980-11-11 Collagen Corporation Non-antigenic collagen and articles of manufacture
US4582640A (en) * 1982-03-08 1986-04-15 Collagen Corporation Injectable cross-linked collagen implant material
US4544516A (en) * 1982-07-28 1985-10-01 Battelle Development Corporation Collagen orientation
US4789663A (en) * 1984-07-06 1988-12-06 Collagen Corporation Methods of bone repair using collagen
US4956178A (en) * 1988-07-11 1990-09-11 Purdue Research Foundation Tissue graft composition
US4902508A (en) * 1988-07-11 1990-02-20 Purdue Research Foundation Tissue graft composition
US5163955A (en) * 1991-01-24 1992-11-17 Autogenics Rapid assembly, concentric mating stent, tissue heart valve with enhanced clamping and tissue alignment
US5420248A (en) * 1991-07-04 1995-05-30 Coletica Unpigmented fish skin, particularly from flat fish, as a novel industrial source of collagen, extraction method, collagen and biomaterial thereby obtained
US5281422A (en) * 1991-09-24 1994-01-25 Purdue Research Foundation Graft for promoting autogenous tissue growth
US5204382A (en) * 1992-02-28 1993-04-20 Collagen Corporation Injectable ceramic compositions and methods for their preparation and use
US5275826A (en) * 1992-11-13 1994-01-04 Purdue Research Foundation Fluidized intestinal submucosa and its use as an injectable tissue graft
US5460962A (en) * 1994-01-04 1995-10-24 Organogenesis Inc. Peracetic acid sterilization of collagen or collagenous tissue
US5948429A (en) * 1994-11-22 1999-09-07 Tissue Engineering, Inc. Methods for preparing biopolymer foams
US5955110A (en) * 1995-04-07 1999-09-21 Purdue Research Foundation, Inc. Multilayered submucosal graft constructs and method for making the same
US5554389A (en) * 1995-04-07 1996-09-10 Purdue Research Foundation Urinary bladder submucosa derived tissue graft
US5885619A (en) * 1995-04-07 1999-03-23 Purdue Research Foundation Large area submucosal tissue graft constructs and method for making the same
US6187047B1 (en) * 1995-10-16 2001-02-13 Orquest, Inc. Bone grafting matrix
US6497875B1 (en) * 1996-04-26 2002-12-24 Case Western Reserve University Multilayer skin or dermal equivalent having a layer containing mesenchymal stem cells
US20040078076A1 (en) * 1996-08-23 2004-04-22 Badylak Stephen F. Purified submucosa graft material
US6666892B2 (en) * 1996-08-23 2003-12-23 Cook Biotech Incorporated Multi-formed collagenous biomaterial medical device
US6206931B1 (en) * 1996-08-23 2001-03-27 Cook Incorporated Graft prosthesis materials
US6793939B2 (en) * 1996-12-10 2004-09-21 Purdue Research Foundation Biomaterial derived from vertebrate liver tissue
US6375989B1 (en) * 1996-12-10 2002-04-23 Purdue Research Foundation Submucosa extracts
US6099567A (en) * 1996-12-10 2000-08-08 Purdue Research Foundation Stomach submucosa derived tissue graft
US6444229B2 (en) * 1998-02-27 2002-09-03 Purdue Research Foundation Submucosa gel compositions
US6264992B1 (en) * 1998-02-27 2001-07-24 Purdue Research Foundation Submucosa as a growth substrate for cells
US6384196B1 (en) * 1998-03-24 2002-05-07 Merck Patent Gesellschaft Process for the preparation of mineralized collagen fibrils and their uses as bone substitute material
US20040037813A1 (en) * 1999-02-25 2004-02-26 Simpson David G. Electroprocessed collagen and tissue engineering
US20050153442A1 (en) * 1999-03-10 2005-07-14 Adam Katz Adipose-derived stem cells and lattices
US6592623B1 (en) * 1999-08-31 2003-07-15 Virginia Commonwealth University Intellectual Property Foundation Engineered muscle
US6592794B1 (en) * 1999-09-28 2003-07-15 Organogenesis Inc. Process of making bioengineered collagen fibrils
US20020170120A1 (en) * 1999-12-07 2002-11-21 Zdenek Eckmayer Method of making a collagen membrane from porcine skin
US6576265B1 (en) * 1999-12-22 2003-06-10 Acell, Inc. Tissue regenerative composition, method of making, and method of use thereof
US20040137616A1 (en) * 2000-02-29 2004-07-15 Isseroff Roslyn R. Corneal epithelial graft composites
US6893812B2 (en) * 2000-05-30 2005-05-17 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Three-dimensional ex vivo angiogenesis system
US20020076816A1 (en) * 2000-12-04 2002-06-20 Jianwu Dai Stem cells and signals developed for use in tissue and organ repair and replacement
US20050019419A1 (en) * 2002-01-11 2005-01-27 Badylak Stephen Francis Biomaterial derived from vertebrate liver tissue
US20060147501A1 (en) * 2003-02-28 2006-07-06 Hillas Patrick J Collagen compositions and biomaterials
US20050014181A1 (en) * 2003-06-18 2005-01-20 Galis Zorina S. Methods and compositions for the assessment of polymer assembly
US20080025956A1 (en) * 2004-02-09 2008-01-31 Indiana University Research And Technology Corporation Blood vessel formation from endothelial colony forming cells
US20060235511A1 (en) * 2004-02-09 2006-10-19 Cook Incorporated Woven implantable device
US20050266556A1 (en) * 2004-02-09 2005-12-01 Yoder Mervin C Isolation, expansion and use of clonogenic endothelial progenitor cells
US20050260748A1 (en) * 2004-02-27 2005-11-24 Michigan State University Adult stem cells and uses thereof
US20050226856A1 (en) * 2004-03-09 2005-10-13 Ahlfors Jan-Eric W Autogenic living scaffolds and living tissue matrices: methods and uses thereof
US20070077652A1 (en) * 2004-09-16 2007-04-05 Tony Peled Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells
US20060165667A1 (en) * 2004-12-03 2006-07-27 Case Western Reserve University Novel methods, compositions and devices for inducing neovascularization
US20070026518A1 (en) * 2005-03-29 2007-02-01 The Regents Of The University Of California Controlling stem cell destiny with tunable matrices
US20090011021A1 (en) * 2005-05-16 2009-01-08 Purdue Research Foundation Engineered Extracellular Matrices
US20070269476A1 (en) * 2006-05-16 2007-11-22 Voytik-Harbin Sherry L Engineered extracellular matrices control stem cell behavior
US20090175922A1 (en) * 2006-05-16 2009-07-09 Voytik-Harbin Sherry L Three dimensional purified collagen matrices
US20080199441A1 (en) * 2006-11-09 2008-08-21 Tony Peled Use of ex-vivo cultured hematopoietic cells for treatment of peripheral vascular diseases
US20090069893A1 (en) * 2007-04-19 2009-03-12 Mikhail Vitoldovich Paukshto Oriented Collagen-Based Materials, Films and Methods of Making Same
US20090280180A1 (en) * 2007-12-10 2009-11-12 Voytik-Harbin Sherry L Collagen-based matrices with stem cells

Cited By (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9446107B2 (en) 2005-12-13 2016-09-20 President And Fellows Of Harvard College Scaffolds for cell transplantation
US11096997B2 (en) 2005-12-13 2021-08-24 President And Fellows Of Harvard College Scaffolds for cell transplantation
US8932583B2 (en) 2005-12-13 2015-01-13 President And Fellows Of Harvard College Scaffolds for cell transplantation
US9132210B2 (en) 2005-12-13 2015-09-15 President And Fellows Of Harvard College Scaffolds for cell transplantation
US10149897B2 (en) 2005-12-13 2018-12-11 President And Fellows Of Harvard College Scaffolds for cell transplantation
US10137184B2 (en) 2005-12-13 2018-11-27 President And Fellows Of Harvard College Scaffolds for cell transplantation
US20070269476A1 (en) * 2006-05-16 2007-11-22 Voytik-Harbin Sherry L Engineered extracellular matrices control stem cell behavior
US9315778B2 (en) 2006-05-16 2016-04-19 Purdue Research Foundation Engineered extracellular matrices control stem cell behavior
US8084055B2 (en) 2006-09-21 2011-12-27 Purdue Research Foundation Collagen preparation and method of isolation
US8512756B2 (en) 2006-09-21 2013-08-20 Purdue Research Foundation Collagen preparation and method of isolation
US9770535B2 (en) 2007-06-21 2017-09-26 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
US20110020216A1 (en) * 2007-06-21 2011-01-27 David James Mooney Scaffolds for cell collection or elimination
US10695468B2 (en) 2007-06-21 2020-06-30 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
US9867905B2 (en) 2007-12-10 2018-01-16 Purdue Research Foundation Collagen-based matrices with stem cells
US20090280180A1 (en) * 2007-12-10 2009-11-12 Voytik-Harbin Sherry L Collagen-based matrices with stem cells
US9821045B2 (en) 2008-02-13 2017-11-21 President And Fellows Of Harvard College Controlled delivery of TLR3 agonists in structural polymeric devices
US10258677B2 (en) 2008-02-13 2019-04-16 President And Fellows Of Harvard College Continuous cell programming devices
US9370558B2 (en) 2008-02-13 2016-06-21 President And Fellows Of Harvard College Controlled delivery of TLR agonists in structural polymeric devices
US10328133B2 (en) 2008-02-13 2019-06-25 President And Fellows Of Harvard College Continuous cell programming devices
US10568949B2 (en) 2008-02-13 2020-02-25 President And Fellows Of Harvard College Method of eliciting an anti-tumor immune response with controlled delivery of TLR agonists in porous polymerlc devices
US9539309B2 (en) 2008-05-30 2017-01-10 President And Fellows Of Harvard College Controlled release of growth factors and signaling molecules for promoting angiogenesis
US9012399B2 (en) 2008-05-30 2015-04-21 President And Fellows Of Harvard College Controlled release of growth factors and signaling molecules for promoting angiogenesis
US20110117170A1 (en) * 2008-05-30 2011-05-19 Lan Cao Controlled Release of Growth Factors and Signaling Molecules for Promoting Angiogenesis
US20120077181A1 (en) * 2009-02-19 2012-03-29 Timo Schmidt Method for the cryopreservation of cells, artificial cell constructs or three-dimensional complex tissues assemblies
US20120122218A1 (en) * 2009-04-13 2012-05-17 President And Fellows Of Harvard College Harnessing Cell Dynamics to Engineer Materials
US9297005B2 (en) * 2009-04-13 2016-03-29 President And Fellows Of Harvard College Harnessing cell dynamics to engineer materials
US20110021753A1 (en) * 2009-07-27 2011-01-27 National Cheng Kung University Method for Preparing Porous Collagen Matrices
US20110020864A1 (en) * 2009-07-27 2011-01-27 National Cheng Kung University Preparation of High Purity Collagen
US8198408B2 (en) 2009-07-27 2012-06-12 National Cheng Kung University Method for preparing porous collagen matrices
US7964704B2 (en) 2009-07-27 2011-06-21 Life Fusion, Llc Preparation of high purity collagen
US10080789B2 (en) 2009-07-31 2018-09-25 President And Fellows Of Harvard College Programming of cells for tolerogenic therapies
US9381235B2 (en) 2009-07-31 2016-07-05 President And Fellows Of Harvard College Programming of cells for tolerogenic therapies
US9610328B2 (en) 2010-03-05 2017-04-04 President And Fellows Of Harvard College Enhancement of skeletal muscle stem cell engraftment by dual delivery of VEGF and IGF-1
US9693954B2 (en) 2010-06-25 2017-07-04 President And Fellows Of Harvard College Co-delivery of stimulatory and inhibitory factors to create temporally stable and spatially restricted zones
US20120027732A1 (en) * 2010-07-27 2012-02-02 Voytik-Harbin Sherry L Thermoreversible collagen
US11202759B2 (en) 2010-10-06 2021-12-21 President And Fellows Of Harvard College Injectable, pore-forming hydrogels for materials-based cell therapies
US9603894B2 (en) 2010-11-08 2017-03-28 President And Fellows Of Harvard College Materials presenting notch signaling molecules to control cell behavior
US10647959B2 (en) 2011-04-27 2020-05-12 President And Fellows Of Harvard College Cell-friendly inverse opal hydrogels for cell encapsulation, drug and protein delivery, and functional nanoparticle encapsulation
US10045947B2 (en) 2011-04-28 2018-08-14 President And Fellows Of Harvard College Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
US9675561B2 (en) 2011-04-28 2017-06-13 President And Fellows Of Harvard College Injectable cryogel vaccine devices and methods of use thereof
US10828337B2 (en) 2011-06-02 2020-11-10 Indiana University Research And Technology Corporation Materials and methods for controlling vasculogenesis from endothelial colony forming cells
US9486512B2 (en) 2011-06-03 2016-11-08 President And Fellows Of Harvard College In situ antigen-generating cancer vaccine
US10406216B2 (en) 2011-06-03 2019-09-10 President And Fellows Of Harvard College In situ antigen-generating cancer vaccine
WO2013013537A1 (en) 2011-07-28 2013-01-31 Wang Shanshan Composite collagen sponge and preparation method thereof
US9439999B2 (en) 2011-07-28 2016-09-13 Harbin Peiqilong Biopharmaceutical Co., Ltd Composite collagen sponge and preparation method thereof
US9937249B2 (en) 2012-04-16 2018-04-10 President And Fellows Of Harvard College Mesoporous silica compositions for modulating immune responses
US11278604B2 (en) 2012-04-16 2022-03-22 President And Fellows Of Harvard College Mesoporous silica compositions comprising inflammatory cytokines comprising inflammatory cytokines for modulating immune responses
US9878071B2 (en) 2013-10-16 2018-01-30 Purdue Research Foundation Collagen compositions and methods of use
US11478574B2 (en) 2013-10-16 2022-10-25 Purdue Research Foundation Collagen compositions and methods of use
US10682400B2 (en) 2014-04-30 2020-06-16 President And Fellows Of Harvard College Combination vaccine devices and methods of killing cancer cells
US11786457B2 (en) 2015-01-30 2023-10-17 President And Fellows Of Harvard College Peritumoral and intratumoral materials for cancer therapy
US11150242B2 (en) 2015-04-10 2021-10-19 President And Fellows Of Harvard College Immune cell trapping devices and methods for making and using the same
US11752238B2 (en) 2016-02-06 2023-09-12 President And Fellows Of Harvard College Recapitulating the hematopoietic niche to reconstitute immunity
US11919941B2 (en) 2016-04-21 2024-03-05 Purdue Research Foundation Cell-collagen-silica composites and methods of making and using the same
US11555177B2 (en) 2016-07-13 2023-01-17 President And Fellows Of Harvard College Antigen-presenting cell-mimetic scaffolds and methods for making and using the same
JP7242553B2 (en) 2017-01-31 2023-03-20 ジェニフィス,インコーポレイテッド Methods and compositions for the preparation of substrates
JP2020505474A (en) * 2017-01-31 2020-02-20 ジェニフィス,エルエルシー Methods and compositions for the preparation of substrates
CN110446500A (en) * 2017-01-31 2019-11-12 格尼菲斯有限责任公司 Method and composition for matrix preparation
WO2018144496A1 (en) * 2017-01-31 2018-08-09 Geniphys, Llc Methods and compositions for matrix preparation
US11739291B2 (en) 2017-04-25 2023-08-29 Purdue Research Foundation 3-dimensional (3D) tissue-engineered muscle for tissue restoration
US11590256B2 (en) * 2017-11-03 2023-02-28 Cellontech Co., Ltd. Medical material produced using collagen and method for producing same
CN115466322A (en) * 2022-09-26 2022-12-13 斐缦(长春)医药生物科技有限责任公司 Self-assembled collagen and preparation method and application thereof

Also Published As

Publication number Publication date
GB0906286D0 (en) 2009-05-20
AU2007297611A1 (en) 2008-03-27
US20120141417A1 (en) 2012-06-07
US8084055B2 (en) 2011-12-27
AU2007297611B2 (en) 2013-02-07
WO2008036393A1 (en) 2008-03-27
GB2455041B (en) 2012-03-07
US8512756B2 (en) 2013-08-20
GB2455041A (en) 2009-06-03
CA2663722A1 (en) 2008-03-27
CA2663722C (en) 2014-12-09

Similar Documents

Publication Publication Date Title
US8512756B2 (en) Collagen preparation and method of isolation
Lee et al. Human hair keratin and its-based biomaterials for biomedical applications
Jha et al. Electrospun collagen: a tissue engineering scaffold with unique functional properties in a wide variety of applications
CA2652138C (en) Three dimensional purified collagen matrices
CA2608708C (en) Engineered extracellular matrix having a specified fibril area fraction
US9315778B2 (en) Engineered extracellular matrices control stem cell behavior
EP2644692B1 (en) High-strength collagen fiber membrane and method for producing same
US20120027732A1 (en) Thermoreversible collagen
WO2009076441A1 (en) Collagen-based matrices with stem cells
JP2017529390A (en) Collagen-based therapeutic delivery system
AU2006247228B2 (en) Engineered extracellular matrices control stem cell behavior
US20120171768A1 (en) Collagen-based matrices for vessel density and size regulation
US8877500B2 (en) Thermally induced gelation of collagen hydrogel and method of thermally inducing gelling a collagen hydrogel
JP2009538854A (en) Isolated natural natural collagen
KR20230091583A (en) Nanofibers for bone regeneration using phycocyanin and fish collagen and preparation method of the same
JP2008512115A (en) Method for isolating biomaterial from tissue and biomaterial extract isolated therefrom
JP2023007495A (en) Cell culture substrate, structure, and structure manufacturing method
GB2463811A (en) Collagen matrices
Onofri Development of an extracellular matrix hydrogel for intestine tissue engineering

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: EXECUTIVE ORDER 9424, CONFIRMATORY LICENSE;ASSIGNOR:PURDUE UNIVERSITY;REEL/FRAME:022382/0419

Effective date: 20090311

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: PURDUE RESEARCH FOUNDATION, INDIANA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VOYTIK-HARBIN, SHERRY L.;KREGER, SETH;BELL, BRETT;AND OTHERS;SIGNING DATES FROM 20120104 TO 20120222;REEL/FRAME:027757/0920

CC Certificate of correction
FPAY Fee payment

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: 7.5 YR SURCHARGE - LATE PMT W/IN 6 MO, LARGE ENTITY (ORIGINAL EVENT CODE: M1555); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FEPP Fee payment procedure

Free format text: 11.5 YR SURCHARGE- LATE PMT W/IN 6 MO, LARGE ENTITY (ORIGINAL EVENT CODE: M1556); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 12TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1553); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 12